US20050119279A1 - Spiro-cyclic compounds useful as anti-inflammatory agents - Google Patents

Spiro-cyclic compounds useful as anti-inflammatory agents Download PDF

Info

Publication number
US20050119279A1
US20050119279A1 US10/957,255 US95725504A US2005119279A1 US 20050119279 A1 US20050119279 A1 US 20050119279A1 US 95725504 A US95725504 A US 95725504A US 2005119279 A1 US2005119279 A1 US 2005119279A1
Authority
US
United States
Prior art keywords
alkyl
heterocyclo
heteroaryl
cycloalkyl
substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/957,255
Other versions
US7199125B2 (en
Inventor
T. G. Dhar
Edwin Iwanowicz
Michele Launay
Dominique Potin
Magali Blandine Maillet
Eric Nicolai
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cerep SA
Bristol Myers Squibb Co
Original Assignee
Cerep SA
Bristol Myers Squibb Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cerep SA, Bristol Myers Squibb Co filed Critical Cerep SA
Priority to US10/957,255 priority Critical patent/US7199125B2/en
Publication of US20050119279A1 publication Critical patent/US20050119279A1/en
Assigned to CEREP SA reassignment CEREP SA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NICOLAI, ERIC, LAUNAY, MICHELE, MAILLET, MAGALI JEANNINE BLANDINE, POTIN, DOMINIQUE
Assigned to BRISTOL-MYERS SQUIBB CO. reassignment BRISTOL-MYERS SQUIBB CO. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: IWANOWICZ, EDWIN, DHAR, T. G. MURALI
Application granted granted Critical
Publication of US7199125B2 publication Critical patent/US7199125B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/20Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains three hetero rings
    • C07D513/20Spiro-condensed systems

Definitions

  • the present invention relates to spiro-cyclic compounds, pharmaceutical compositions containing them, and methods of using such compounds in treating inflammatory or immune disease.
  • a key event in an immune response involves the migration of leukocytes to a disease site. During an inflammatory response, leukocytes are recruited to the site of injury and extravasated by a series of cellular interactions involving cell-cell and cell-substrate adhesion.
  • Integrins are expressed on cell surfaces and function in cell-cell and cell-substrate adhesion. Integrins are alpha-beta heterodimers: each integrin has an alpha ( ⁇ ) subunit non-covalently binded to a beta ( ⁇ ) subunit. When activated, integrins bind to extracellular ligands and induce adhesion (the expression of integrins on a cell surface alone is inadequate for adhesion—they must be activated to become adhesive).
  • the integrin activation state is transient, such that there is a rapid flux between adhesive and non-adhesive states which is important for cell movement, e.g., a cell is endowed with the ability to rapidly adhere to various cell surfaces and matrices and to migrate among cells and tissue.
  • integrins having a ⁇ 2 or CD18 subunit which comprise the CD11/CD18 integrin sub-family, namely, Lymphocyte Function-associated Antigen 1 (LFA-1) (CD11a/CD18 or ⁇ L ⁇ 2 ); Macrophage Antigen 1 (Mac-1) (CD11b/CD18 or (XMP2); p150,95 (CD11c/CD18 or (XP2); and ⁇ D ⁇ 2 .
  • LFA-1 Lymphocyte Function-associated Antigen 1
  • Mac-1 CD11b/CD18 or (XMP2)
  • p150,95 CD11c/CD18 or (XP2)
  • ⁇ D ⁇ 2 The CD11/CD18 family of integrins is also referred to as Leukointegrins as they are expressed on the surface of various leukocyte cells, and they mediate a number of inflammation-related cellular interactions. See Diamond et al., “ The Dynamic Regulation of Integrin Adhesiveness,” Current Biology, Vol. 4 (1994
  • Ligands to LFA-1 and Mac-1 comprise the intercellular adhesion molecule (ICAM) ICAM-1.
  • the primary CD11/CD18 integrin is LFA-1, which also binds with ICAM-2 and ICAM-3.
  • ICAMs are found on endothelium cells, leukocytes, and other cell types, and their interaction with CD11/CD18 integrins is critical to immune system function.
  • the interaction between the CD18 integrins, particularly LFA-1, and ICAMs mediates antigen presentation, T-cell proliferation, and adhesion between the endothelium and activated leukocytes which is necessary for leukocytes to migrate from the circulatory system into tissue.
  • Leukocyte Adhesion Deficiency A condition termed “Leukocyte Adhesion Deficiency” has been identified in patients having a deficiency in CD18 integrins. These patients are unable to mount a normal inflammatory or immune response; they suffer from disorders such as recurrent infections, poor wound healing, granulocytosis, progressive periodontitis, and umbilical cord separation. See Anderson et al., “ Leukocyte LFA -1, OKMI, p150,95 Deficiency Syndrome: Functional and Biosynthesis Studies of Three Kindreds,” Fed. Proc., Vol. 44 (1985), at pp. 2671-2677.
  • LFA-1 lipid-binding protein-1
  • Blocking LFA-1 reportedly inhibits the influx of leukocytes in almost every system, including the skin, peritoneum, synovium, lung, kidney, and heart, and blocking ICAM-1 would be expected to have similar effects.
  • present therapies for many inflammatory and immune diseases have drawbacks. For example, current treatments for asthma include ⁇ 2 -agonists, inhaled corticosteroids, and LTD 4 antagonists. However, ⁇ 2 -agonists have limited efficacy and inhaled corticosteroids raise safety concerns.
  • current therapies include PUVA, methotrexate, cyclosporin A, and topical treatments.
  • the first three of these therapies raise toxicity issues over long-term (6-9 month) use, whereas topical treatments have limited efficacy. Additionally, these treatments typically are applied only in response to flares and not as a prophylaxis measure.
  • LFA-1/ICAM Compounds that reportedly inhibit LFA-1/ICAM for use as anti-inflammatory agents include thiadiazole-based compounds (see Intern. Pub. No. WO 99/20,618, “ Thiadiazole Amides Useful as Anti-Inflammatory Agents ” filed by Pharmacia & Upjohn Co.; and WO 99/20,617, also to Pharmacia and Upjohn); and thiazole compounds linked to phenyl and pyrazole rings (Sanfilippo et al., “ Novel Thiazole Based Heterocyclos as Inhibitors of LFA -1 /ICAM - J Mediated Cell Adhesion,” J. Med. Chem., Vol. 38 (1995) at pp.1057-1059).
  • Small molecules that reportedly are antagonists to the binding of ICAMs with CD18 integrins include various benzylamines and 2-bromobenzoyltryptophan compounds (see Intern. Pub. No. WO99/49,856, “ Antagonists for Treatment of CD 11 /CD 18 Adhesion Receptor Mediated Disorders,” filed by Genentech, Inc.), and 1-(3,5 dichlorophenyl) imidazolidines (see Intern. Pub. No. WO98/39303, “ Small Molecules Useful in the Treatment of Inflammatory Disease,” filed by Boehringer Ingelheim Pharmaceuticals, Inc.
  • Hydantoin compounds are disclosed in WO 01/30781 A2 (published May 3, 2001) to Tanabe Seiyaku Co. Ltd, WO 03029245 A1 (published Apr. 10, 2003) to Bristol-Myers Squibb; and WO 01030781 A1 (published May 3, 2003) to Bristol-Myers Squibb.
  • Diazaspiroheptane compounds are disclosed in Park et al., “ Preparation of a 990 Member Chemical Compound Library of Hydantoin and Isoxazoline - Containing Heterocyclos Using Multipin Technology,” J. Comb. Chem., Vol. 3(2) (2001), at pp.
  • the present invention provides compounds useful in treating inflammatory or immune disease having the formula (I):
  • the present invention is also directed to pharmaceutical compositions useful in treating immune or inflammatory diseases comprising compounds of formula (I), or stereoisomers, pharmaceutically-acceptable salts, pharmaceutically-acceptable carriers or diluents thereof.
  • the invention further relates to methods of treating immune or inflammatory diseases comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound according to formula (I).
  • alkyl and alk refers to a straight or branched chain alkane (hydrocarbon) radical containing from 1 to 12 carbon atoms, preferably 1 to 6 carbon atoms.
  • exemplary “alkyl” groups include methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, isobutyl pentyl, hexyl, isohexyl, heptyl, 4,4-dimethylpentyl, octyl, 2,2,4-trimethylpentyl, nonyl, decyl, undecyl, dodecyl, and the like.
  • C 1 -C 6 alkyl refers to a straight or branched chain alkane (hydrocarbon) radical containing from 1 to 6 carbon atoms, such as methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, and isobutyl.
  • Substituted alkyl refers to an alkyl group substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment.
  • substituents include but are not limited to one or more of the following groups: hydrogen, halogen (e.g., a single or multiple halo substitutents) trifluoromethyl, trifluoromethoxy, cyano, nitro, alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocyclo, aryl, OR a , SR a , S( ⁇ O)R e , S( ⁇ O) 2 R e , P( ⁇ O) 2 R e , S( ⁇ O) 2 OR e , P( ⁇ O) 2 OR e , NR b R c , NR b S( ⁇ O) 2 R e , NR b P( ⁇ O) 2 R e , S( ⁇ O) 2 R
  • substituents also include spiro-attached or fused cylic substituents, especially spiro-attached cycloalkyl, spiro-attached heterocyclo, fused cycloalkyl, fused heterocyclo, fused aryl, or fused heteroaryl,
  • groups such as alkyl, cycloalkyl, alkenyl, alkynyl, cycloalkenyl, heterocyclo, aryl and heteroaryl can themselves be optionally substituted, as described infra.
  • alkenyl refers to a straight or branched chain hydrocarbon radical containing from 2 to 12 carbon atoms and at least one carbon-carbon double bond. Exemplary such groups include ethenyl or allyl. “Substituted alkenyl” refers to an alkenyl group substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment. Exemplary substituents include, but are not limited to, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents.
  • alkynyl refers to a straight or branched chain hydrocarbon radical containing from 2 to 12 carbon atoms and at least one carbon to carbon triple bond. Exemplary such groups include ethynyl. “Substituted alkynyl” refers to an alkynyl group substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment. Exemplary substituents include, but are not limited to, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents.
  • halo or “halogen” refers to chloro, bromo, fluoro and iodo.
  • haloalkyl means a substituted alkyl having one or more halo substituents.
  • haloalkyl includes mono, bi, and trifluoromethyl.
  • haloalkoxy means an alkoxy group having one or more halo substituents.
  • haloalkoxy includes OCF 3 .
  • cycloalkyl refers to fully saturated and partially unsaturated hydrocarbon rings of 3 to 9, preferably 3 to 7 carbon atoms.
  • cycloalkyl includes such rings Exemplary such groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, etc.
  • Cycloalkyl groups may be unsubstituted or substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment.
  • substituents include, but are not limited to, nitro, cyano, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents.
  • substituents also include spiro-attached or fused cylic substituents, especially spiro-attached cycloalkyl, spiro-attached heterocyclo, fused cycloalkyl, fused cycloalkenyl, fused heterocyclo, fused aryl, or fused heteroaryl, the aforementioned cycloalkyl, heterocyclo, aryl and heteroaryl can themselves be optionally substituted.
  • aryl refers to unsubstituted or substituted cyclic, aromatic hydrocarbon groups which have 1 to 5 aromatic rings, especially monocyclic or bicyclic groups such as phenyl, biphenyl or naphthyl. Where containing two or more aromatic rings (bicyclic, etc.), the aromatic rings of the aryl group may be joined at a single point (e.g., biphenyl), or fused (e.g., naphthyl, phenanthrenyl and the like). The aryl group may be substituted by one or more substituents, preferably 1 to 3 substituents, at any point of attachment.
  • substituents include, but are not limited to, nitro, cycloalkyl, cyano, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents.
  • substituents also include fused cylic, especially fused cycloalkyl, fused heterocyclo, fused aryl, or fused heteroaryl.
  • heterocyclo refers non-aromatic 3 to 7 membered monocyclic groups, 7 to 11 membered bicyclic groups, and 10 to 15 membered tricyclic groups, in which at least one of the rings has at least one heteroatom (O, S or N).
  • Each heerocyclo group may be unsubstituted or substituted.
  • Each ring of the heterocyclo group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less, and further provided that the ring contains at least one carbon atom.
  • the fused rings completing bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated.
  • the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quaternized.
  • the heterocyclo group may be attached at any available nitrogen or carbon atom in any fashion, including spiro- attachments.
  • the heterocyclo group may be substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment.
  • substituents include, but are not limited to, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, nitro, oxo (i.e., ⁇ O), cyano, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents.
  • substituents also include spiro-attached or fused cylic substituents at any available point or points of attachment, especially spiro-attached cycloalkyl, spiro-attached cycloalkenyl, spiro-attached heterocyclo, fused cycloalkyl, fused cycloalkenyl, fused heterocyclo, fused aryl, or fused heteroaryl, the aforementioned cycloalkyl, cycloalkenyl, heterocyclo, aryl and heteroaryl can themselves be optionally substituted.
  • Exemplary monocyclic heterocyclo groups include azetidinyl, pyrrolidinyl, oxetanyl, imidazolinyl, oxazolidinyl, isoxazolinyl, thiazolidinyl, isothiazolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolodinyl, 2-oxoazepinyl, azepinyl, 4-piperidonyl, tetrahydropyranyl, morpholinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, 1,3-dioxolane and tetrahydro-1,1-dioxothienyl and the like.
  • Exemplary bicyclic heterocyclo groups include quinuclidin
  • heteroaryl refers to substituted and unsubstituted aromatic 5 or 6 membered monocyclic groups, 9 or 10 membered bicyclic groups, and 11 to 14 membered tricyclic groups which have at least one heteroatom (O, S or N) in at least one of the rings.
  • Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom.
  • the fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated.
  • the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quatemized.
  • Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non-aromatic.
  • the heteroaryl group may be attached at any available nitrogen or carbon atom of any ring. “Substituted heteroaryl” refers to heteroaryl groups substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment.
  • substituents include, but are not limited to, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, nitro, cyano, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents.
  • substituents also include or fused cylic substituents at any available points of attachment, especially fused cycloalkyl, fused cycloalkenyl, fused heterocyclo, fused aryl and fused heteroaryl, the aforementioned cycloalkyl, cycloalkenyl, heterocyclo, aryl and heteroaryl can themselves be optionally substituted.
  • Exemplary monocyclic heteroaryl groups include pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl and the like.
  • Exemplary bicyclic heteroaryl groups include indolyl, benzothiazolyl, benzodioxolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl, dihydroisoindolyl, tetrahydroquinolinyl and the like.
  • Exemplary tricyclic heteroaryl groups include carbazolyl, benzidolyl, phenanthrollinyl, acridinyl, phenanthridinyl, xanthenyl and the like.
  • aryl e.g., phenyl
  • cycloalkyl e.g., cyclohexyl
  • heterocyclo e.g., pyrrolidinyl
  • heteroaryl e.g., imidazolyl
  • the reference is intended to include rings having 0 to 3, preferably 0-2, substituents selected from those recited above for the aryl, cycloalkyl, heterocyclo and/or heteroaryl groups, as appropriate.
  • heteroatoms shall include oxygen, sulfur and nitrogen.
  • carbocyclic means a saturated or unsaturated monocyclic or bicyclic ring in which all atoms of all rings are carbon. Thus, the term includes cycloalkyl and aryl rings. The carbocyclic ring may be substituted in which case the substituents are selected from those recited above for cycloalkyl and aryl groups.
  • the ring or group may be fully unsaturated or partially unsaturated.
  • protecting groups for the methods and compounds described herein include, without limitation, those described in standard textbooks, such as Greene, T. W. et al., Protective Groups in Organic Synthesis, Wiley, N.Y. (1999).
  • any heteroatom with unsatisfied valences is assumed to have hydrogen atoms sufficient to satisfy the valences.
  • salts form salts which are also within the scope of this invention.
  • Reference to a compound of the formula I herein is understood to include reference to salts thereof, unless otherwise indicated.
  • zwitterions inner salts may be formed and are included within the term “salt(s)” as used herein.
  • Salts of the compounds of the formula I may be formed, for example, by reacting a compound I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
  • the compounds of formula I which contain a basic moiety may form salts with a variety of organic and inorganic acids.
  • Exemplary acid addition salts include acetates (such as those formed with acetic acid or trihaloacetic acid, for example, trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, hydroxyethane
  • the compounds of formula I which contain an acidic moiety may form salts with a variety of organic and inorganic bases.
  • Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as benzathines, dicyclohexylamines, hydrabamines (formed with N,N-bis(dehydroabietyl) ethylenediamine), N-methyl-D-glucamines, N-methyl-D-glycamides, t-butyl amines, and salts with amino acids such as arginine, lysine and the like.
  • Basic nitrogen-containing groups may be quaternized with agents such as lower alkyl halides (e.g. methyl, ethyl, propyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g. dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (e.g. decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides), aralkyl halides (e.g. benzyl and phenethyl bromides), and others.
  • lower alkyl halides e.g. methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates e.g. dimethyl, diethyl, dibutyl, and diamyl sulfates
  • Prodrugs and solvates of the compounds of the invention are also contemplated herein.
  • the term “prodrug” as employed herein denotes a compound which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of the formula I, or a salt and/or solvate thereof.
  • Solvates of the compounds of formula I include, for example, hydrates.
  • stereoisomers of the present compounds are contemplated within the scope of this invention.
  • Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers (e.g., as a pure or substantially pure optical isomer having a specified activity), or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
  • the chiral centers of the present invention may have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • racemic forms can be resolved by physical methods, such as, for example, fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography.
  • the individual optical isomers can be obtained from the racemates by any suitable method, including without limitation, conventional methods, such as, for example, salt formation with an optically active acid followed by crystallization.
  • Preferred compounds of the invention include compounds of formula (I) in which one or more, preferably all, of the following substituents are as defined below:
  • Particularly preferred compounds, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, within the scope of formula (I) are those wherein Q-Ar together form:
  • preferred compounds of the invention include compounds of forula (Ia)
  • Particularly preferred compounds within the scope of formula (Ia), or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, are those in which one or more, preferably all, of the following substituents re as defined below:
  • preferred compounds of the invention include compounds of formula (Ib)
  • Particularly preferred compounds within the scope of formula (Ib), or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, are those in which one or more, preferably all, of the following substituents are as defined below:
  • Especially preferred compounds are those having the formula, (i) (2R,5S,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
  • the compounds of the present invention may be prepared by methods such as those illustrated in the following schemes. Solvents, temperatures, pressures, and other reaction conditions may readily be selected by one of ordinary skill in the art. Starting materials are commercially available or readily prepared by one of ordinary skill in the art. The symbols used in the followoing schemes are defined as above, unless otherwise indicated.
  • Compounds of the general formula (Ia) may be obtained from compounds of formula (1) by dehydrative cyclization. The choice of the dehydrating agent will be readily apparent to one skilled in the art of organic chemistry. Such methods are, for example described by Greene, Theodora W. and Wuts, Peter G. M.
  • compounds of formula (Ia) may be obtained by refluxing (1) and (2) in a solvent such as xylene, a catalytic amount of acid such a para-toluenesulfonic acid and at a temperature range of 100-140° C. with the azeotropic removal of water.
  • compounds of formula (Ia) may be obtained for example by stirring (1) and (2) in a solvent such as ethanol at a temperature range of 20-50° C. (Refouvelet, Bernard et al.: Chem.Pharm.Bull., 1994, 42 (5), 1076-1083)
  • Compounds of the general formula (1) may be obtained by the oxidation of compounds of general formula (3) as shown in Scheme 2.
  • the choice of the oxidizing agent will be readily apparent to one skilled in the art of organic chemistry. Such methods are, for example described by Hudlicky, M., in “oxidations in organic chemistry”, ACS monograph 186 (1990), American Chemical Society, Washington, D.C.
  • compounds of formula (1) may be obtained from compounds of formula (3) by using an oxidizing agent such as pyridinium chlorochromate, in a solvent like dichloromethane at temperatures ranging from 0° C. to 40° C.
  • Compounds of formula (3) can be prepared by reacting the proline derivative (7) with an isocyanate (R 1 NCO) in DMF in the presence of K 2 CO 3 . See Issartel et al., Eur. J. Med. Chem. Chim. Ther., Vol. 31; 9 (1996), at pp. 717-724, incorporated herein by reference.
  • the isocyantes useful for the preparation of compounds to this invention may be commercially available or readily prepared by many methods known to one skilled in the art of organic chemistry.
  • Compounds of formula (5) can be prepared from compounds of formula (4) by cyclizing the proline compound of formula (4) with anhydrous chloral in acetonitrile. See Orsini et al., J. Heterocyclic Chem., (1989), pp. 837-841, incorporated herein by reference.
  • Compounds of formula (3) can be prepared from compounds of formula (9) by reacting (9) with BrCH 2 —R 4 in the presence of LiHMDS or LDA and THF. See Kobayashi et al., Bull. Chem. Soc. Jpn., Vol. 67; 11 (1994), at pp. 3082-3087, incorporated herein by reference.
  • the protecting group on the oxygen can then be deprotected by one skilled in the art of organic chemistry. Such methods are, for example described by Greene, Theodora W. and Wuts, Peter G. M. in “Protective groups in organic synthesis” 3 rd Ed., (1999) Publisher (John Wiley and Sons, Inc., New York, N.Y).
  • Compounds of formula (9) can be prepared from compounds of formula (8) by reacting proline derivatives (8) with isocyanate R 2 NCO in an aqueous basic media, then cyclizing the intermediate ureidoacid in refluxing toluene in the presence of a catalytic amount of sulfuric acid. See French patent No. FR230083 and Great Britain Pat. No. B 1 503 244, titled “1,5-Alkylene-3-aryl Hydantoin Derivatives,” both to Mitsubishi Chemical Indus., Inc., incorporated herein.
  • the compounds and compositions of this invention are antagonists of LFA-1, Mac-1, and/or ICAMs. Thus, they are useful in treating various inflammatory diseases and disorders associated with the action of LFA-1, Mac-1, and/or ICAMs, particularly LFA-1:ICAM-1.
  • LFA-1:ICAM-1 LFA-1:ICAM-1.
  • the term “Leukointegrin/ICAM-associated condition” is used herein for ease of reference to refer to those diseases or disorders that are associated with the action or levels of LFA-1, Mac-1 and/or ICAM-1, ICAM-2, or ICAM-3.
  • treating includes prophylactic and therapeutic uses and refers to the alleviation of symptoms of a particular disorder in a patient, the improvement of an ascertainable measurement associated with a particular disorder, or the prevention of a particular immune response (such as transplant rejection).
  • patient refers to a mammal, preferably a human.
  • inventive compounds and compositions are useful for treating a wide range of conditions, as the action of LFA-1 and/or ICAMs is associated with the influx of leukocytes in almost every system, including the skin, peritoneum, synovium, lung, kidney and heart.
  • the inventive compounds may be used to treat conditions resulting from a response of the specific immune system in a patient or the nonspecific immune system.
  • Such conditions include, for example, graft vs host reactions and transplant rejection (e.g., kidney, liver, heart, lung, pancreas, bone marrow, cornea, small bowel, skin allografts, skin homografts and heterografts, etc.); psoriasis, organ-tissue autoimmune diseases (e.g., Raynaud's syndrome), autoimmune thyroiditis, dermatitis, multiple sclerosis, rheumatoid arthritis, insulin dependent diabetes mellitus, uveitis, inflammatory bowel disease including Crohn's disease and ulcerative colitis and systemic lupus erythematosus, adult respiratory distress syndrome, shock, oxygen toxicity, multiple organ injury syndrome secondary to septicemia, multiple organ injury syndrome secondary to trauma, reperfusion injury of tissue due to cardiopulmonary bypass, myocardial infraction or use with thrombolysis agents, acute glomerulonephritis, vasculitis, reactive arthritis, dermatosis with acute inflammatory components
  • the compounds of the present invention also may be used to treat allergic conditions such as eczema and asthma or as an adjunct to minimize toxicity with cytokine therapy in the treatment of cancers.
  • the compounds may be used to treat inflammatory conditions that involve the infiltration of T-cells and chronic inflammatory responses, hypersensitivity reactions, such as skin hypersensitivity reactions (including poison ivy and poison oak), immune responses associated with delayed hypersensitivity mediated by cytokines and T-lymphocytes, and metastases.
  • the compounds of this invention further have utility in treating hypogonadism, frailty, osteoporosis, sexual dysfunction, wasting, such as wasting syndromes associated with cancer and AIDS, and anemia.
  • the compounds further have utility in treating cancers, including but not limited to cancers of the breast, brain, skin, ovary, endometrium, bladder, prostate, lung, colon, lymphatic system, liver and kidney.
  • the inventive compounds are useful for conditions such as hirsutism, Alzheimer's disease, non-insulin dependent diabetes mellitus, acne, seborrhea, alopecia, fibroids, hyperpilosity, cachexia, polycystic ovarian syndrome, anorexia, contraception, drug withdrawal syndrome, pregnancy termination, and benign prostate hypertrophy.
  • the compounds are further useful as antiangiogenic agents. Additionally, the compounds may be useful as inhibitors of protein prenyltransferases, particularly famesyltransferase and the prenylation of the oncogene protein Ras. As such, the inventive compounds may potentially be useful for treating and/or preventing the diseases and disorders referred to in WO 01/45704, which is incorporated herein by reference.
  • the compounds When used as anti-inflammatory agents, the compounds may be administered prior to the onset of, at, or after the initiation of inflammation.
  • the compounds When used prophylactically, the compounds are preferably provided in advance of any inflammatory response or symptom (for example, prior to, at, or shortly after the time of an organ or tissue transplant but in advance of any symptoms or organ rejection). Administration of the compounds may prevent or attenuate inflammatory responses or symptoms.
  • the present invention thus provides methods for treating such conditions as those listed above, comprising administering to a subject in need thereof an effective amount of at least one compound of formula (I) or a salt thereof.
  • Other therapeutic agents such as those described below may be employed in combination with the compounds of formula (I).
  • such other therapeutic agent(s) may be administered prior to, simultaneously with, or following the administration of the inventive compounds.
  • the present invention also provides pharmaceutical compositions capable of treating the Leukointegrin/ICAM-associated conditions and above-described diseases and disorders.
  • inventive compositions may contain other therapeutic agents and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulation.
  • the compounds of formula (I) may be administered by any means suitable for the condition to be treated, which may depend on the need for site-specific treatment or quantity of drug to be delivered. Topical administration is generally preferred for skin-related diseases, and systematic treatment preferred for cancerous or pre-cancerous conditions, although other modes of delivery are contemplated.
  • the compounds may be delivered orally, such as in the form of tablets, capsules, granules, powders, or liquid formulations including syrups; topically, such as in the form of solutions, suspensions, gels or ointments; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrastemal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; rectally such as in the form of suppositories; or liposomally.
  • subcutaneous, intravenous, intramuscular, or intrastemal injection or infusion techniques e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions
  • nasally such as by inhalation spray
  • topically such as in the form of a cream or ointment
  • rectally
  • Dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents may be administered.
  • the compounds may be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved with suitable pharmaceutical compositions or, particularly in the case of extended release, with devices such as subcutaneous implants or osmotic pumps.
  • compositions for topical administration include a topical carrier such as PLASTIBASE® (mineral oil gelled with polyethylene).
  • compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art.
  • the inventive compounds may also be orally delivered by sublingual and/or buccal administration, e.g., with molded, compressed, or freeze-dried tablets.
  • compositions may include fast-dissolving diluents such as mannitol, lactose, sucrose, and/or cyclodextrins.
  • fast-dissolving diluents such as mannitol, lactose, sucrose, and/or cyclodextrins.
  • high molecular weight excipients such as celluloses (AVICEL®) or polyethylene glycols (PEG); an excipient to aid mucosal adhesion such as hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), sodium carboxymethyl cellulose (SCMC), and/or maleic anhydride copolymer (e.g., GANTREZ®); and agents to control release such as polyacrylic copolymer (e.g., CARBOPOL 934®).
  • Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • compositions for nasal aerosol or inhalation administration include solutions which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance absorption and/or bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
  • compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • suitable non-toxic, parenterally acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • compositions for rectal administration include suppositories which may contain, for example, suitable non-irritating excipients, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures but liquefy and/or dissolve in the rectal cavity to release the drug.
  • suitable non-irritating excipients such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures but liquefy and/or dissolve in the rectal cavity to release the drug.
  • the effective amount of a compound of the present invention may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for a patient of from about 0.05 to 100 mg/kg of body weight of active compound per day, which may be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day. It will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors, including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and the particular condition sought to be treated and its severity.
  • Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats, horses, and the like, subject to Leukointegrin/ICAM associated conditions and/or subject to any of the above-referenced diseases and disorders.
  • inventive compounds and compositions may be employed alone or in combination with each other and/or other suitable therapeutic agents useful in treating Leukointegrin/ICAM-associated conditions and diseases and disorders referenced above.
  • suitable therapeutic agents include corticosteroids, cyclosporin, methotrexate; CELLCEPTTM (mycophenolate mofetil), co-stimulation blockades, growth hormones, and growth hormone secretagogues.
  • inventive compounds may be administered either alone or in combination with anti-cancer and cytotoxic agents and treatments useful in treating cancer or other proliferative diseases, for example, where the second drug has the same or different mechanism of action than the present compounds.
  • Examples of classes of anti-cancer and cytotoxic agents useful in combination with the present compounds include but are not limited to: alkylating agents such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; antimetabolites such as folate antagonists, purine analogues, and pyrimidine analogues; antibiotics such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; enzymes such as L-asparaginase; famesyl-protein transferase inhibitors; hormonal agents such as glucocorticoids, estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, octreotide acetate; microtubule-disruptor agents, such as ecteinascidins or their analogs and derivatives; microtubule-
  • paclitaxel paclitaxel
  • carboplatin doxorubicin
  • carmninomycin daunorubicin
  • idarubicin aminopterin
  • methotrexate methopterin
  • mitomycin C ecteinascidin 743
  • porfiromycin 5-fluorouracil
  • 6-mercaptopurine gemcitabine
  • cytosine arabinoside podophyllotoxin or podophyllotoxin derivatives
  • etoposide etoposide phosphate or teniposide
  • melphalan vinblastine, vincristine, leurosidine, vindesine and leurosine
  • vinblastine vincristine
  • leurosidine leurosidine
  • vindesine and leurosine mechlorethamine hydrochloride
  • cyclophosphamhide chlorambucil, ifosfamide, busulfan, carmustin, lomustine
  • anticancer and other cytotoxic agents examples include the following: epothilone derivatives as found in German Patent No. 4138042.8; WO 97/19086, WO 98/22461, WO 98/25929, WO 98/38192, WO 99/01124, WO 99/02224, WO 99/02514, WO 99/03848, WO 99/07692, WO 99/27890, WO 99/28324, WO 99/43653, WO 99/54330, WO 99/54318, WO 99/54319, WO 99/65913, WO 99/67252, WO 99/67253 and WO 00/00485; cyclin dependent kinase inhibitors as found in WO 99/24416; and prenyl-protein transferase inhibitors as found in WO 97/30992 and WO 98/54966.
  • inventive compounds have been tested in cell-cell assays and demonstrated activity consistent with inhibition of LFA-1 and/or ICAM-1.
  • H1-Hela cells were released from their growth flask using versene (Gibco, Grand Island, N.Y.). Following centrifugation, the cells were resuspended in growth medium: DMEM (Gibco), 10% fetal calf serum (Hyclone, Logan, Utah), 1% Pen-Strep (Gibco), and 1% L-glutamine (Gibco) and plated for growth at 5,000 cells/well in a 96-well plate.
  • DMEM Gibco
  • 10% fetal calf serum Hyclone, Logan, Utah
  • Pen-Strep Gibco
  • L-glutamine 1% L-glutamine
  • HSB-2 cells were divided to 2 ⁇ 10 5 /ml in growth medium: RPMI 1640 (Gibco), 10% FCS, 1% Pen-Strep, and 1% L-glutamine.
  • RPMI 1640 Gibco
  • FCS fetal bovine serum
  • Pen-Strep 1% Pen-Strep
  • L-glutamine 1% L-glutamine.
  • day #3 the cells were centrifuged at 534 ⁇ G for 8 minutes, washed, and resuspended in HBSS at 5 ⁇ 10 7 /ml.
  • Calcein-AM, 10 ⁇ M (Molecular Probes, Eugene, Oreg.) and 100 nM phorbol myristate acetate (SIGMA, St. Louis, Mo.) were added to the labeling and activation mix.
  • SIGMA nM phorbol myristate acetate
  • the medium was aspirated from the H1-HeLa cells and the plates washed once with HBSS, followed by the addition of 50 ⁇ l of HBSS. An additional 50 ⁇ l of HBSS containing compound solution, DMSO, or anti-CD18 antibody was then added to each well.
  • To the H1-HeLa cells were added 200,000 HSB-2 cells/well in 100 ⁇ l, followed by incubation in the dark for 30 minutes. The wells were then washed three times to remove the unbound cells. A fluorescence plate reader was then used to determine the number of bound HSB-2 cells. The percent inhibition due to the compound was calculated using the vehicle control as 0% inhibition and the antibody blocked adhesion as 100% inhibition.
  • human umbilical vein endothelial cells (HUVEC) (passage 3, Clonetics, San Diego, Calif.) were placed into a T-75 flask containing EGM bulletkit media (Clonetics) for growth.
  • 96-well tissue culture plates were coated with 100 ⁇ l/well of 2.5 ⁇ g/ml mouse Type IV collagen (Gibco) diluted in 0.1 M acetic acid. Following incubation for at least three hours, the collagen was removed and the plate washed three times with HBSS (Gibco). The HUVEC flask was trypsinized, and HUVEC were plated on the collagen coated wells at 1250 cells/200 ⁇ l/well for use four days later. Twenty hours prior to use, the medium was removed and cells were stimulated with 200 ⁇ l of 1 ⁇ g/ml lipopolysaccharide (LPS, Sigma, St. Louis, Mo.) in EGM.
  • LPS lipopolysaccharide
  • the LPS-containing medium was removed, the wells were washed with HBSS, and 50 ⁇ l of HBSS was added to the wells. An additional 50 ⁇ l containing compound solution, DMSO or blocking anti-CD18 was then added to each well.
  • HSB-2 cells were then divided to 2 ⁇ 10 5 /ml in RPMI 1640 (Gibco), 10% FCS (Hyclone, Logan, Utah), 1% Pen-Strep (Gibco), and 1% L-glutarmne (Gibco). The following day, the cells were centrifuged at 534 ⁇ G for 8 minutes, washed, and resuspended in HBSS at 5 ⁇ 10 7 /ml. For activation and labeling, calcein-AM, 10 ⁇ M (Molecular Probes, Eugene, Oreg.) and 100 nM phorbol myristate acetate (Sigma, St. Louis, Mo.) were added and the cells incubated at 37° C. for 30 minutes. Following the addition of ten ml of HBSS, the cells were centrifuged, resuspended, and counted.
  • HUVEC cells To the HUVEC cells were added 200,000 labeled and activated HSB-2 cells/well in 100 ⁇ l, followed by incubation in the dark for 30 minutes. To remove unbound cells, the wells were washed three times with HBSS. A fluorescence plate reader was used to determine the number of HSB-2 cells bound. The percent inhibition due to the compound was calculated with the vehicle control set at 0% inhibition and the antibody-blocked adhesion set at 100% inhibition.
  • the reaction mixture was cooled to room temperature, concentrated under reduced pressure and partitioned between dichloromethane (25 mL) and IN sodium hydroxide (10 mL).
  • the dichloromethane layer was dried over sodium sulfate, concentrated and purified by silica gel chromatography using hexane/ethyl acetate as the eluent.
  • Isomer 1 (2R,5S,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione (35 mg).
  • reaction mixture was purged with nitrogen and heated at 80° C. for eighteen hours, cooled to room temperature and filtered over a thin pad of celite.
  • reaction mixture was cooled to room temperature, diluted with t-butyl methyl ether, washed with water, NaHCO 3 solution, water then dried over sodium sulfate, concentrated and purified by silica gel chromatography using dichloromethane/acetone as the eluent.
  • Isomer 2 (ethyl (4′S,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate):
  • reaction mixture was cooled to room temperature, diluted with t-butyl methyl ether, washed with water, NaHCO 3 solution, water then dried over sodium sulfate, concentrated to give an oil (0.40 g), (0.10 g) of which was purified by silica gel chromatography using dichloromethane/acetone as the eluent.
  • Isomer 1 (ethyl (4′R,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate):
  • Isomer 2 (ethyl (4′R,6S,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate):
  • the reaction mixture was concentrated under reduced pressure and partitioned between dichloromethane (25 mL) and 1N Hydrochloric acid (10 mL).
  • the dichloromethane layer was washed with 1N Sodium Bicarbonate, water then dried over sodium sulfate, concentrated and purified by silica gel chromatography using cyclohexane/ethyl acetate as the eluent, diluted with t-butyl methyl ether, washed with water, NaHCO 3 solution, water then dried over sodium sulfate, concentrated and purified by silica gel chromatography using dichloromethane/acetone as the eluent. yield: 0.016 g.

Abstract

The present invention is directed to compounds having the formula (I):
Figure US20050119279A1-20050602-C00001

useful in treating inflammatory and immune diseases, in which K and L are independently, O or S; Q is —C(═O)— or optionally substituted C1-6alkylene; Ar is optionally-substituted aryl or heteroaryl; J1, J2, J3 and Y are selected so that ring A is a five-to-six membered optionally-substituted cycloalkenyl or heterocyclo ring having 0 to 2 nitrogen heteroatoms; R1 is N or C(R9); and R2, and R3, are as defined in the specification.

Description

  • This application claims the benefit of priority from U.S. Provisional Application Ser. No. 60/508,165 filed Oct. 2, 2003, the entirety of which is incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to spiro-cyclic compounds, pharmaceutical compositions containing them, and methods of using such compounds in treating inflammatory or immune disease.
  • BACKGROUND OF THE INVENTION
  • Cells adhere to other cells and to substrates through specific, regulated processes that are critical to various biological functions. The proper functioning of the immune system is dependent upon adhesive interactions and cell migration. A key event in an immune response involves the migration of leukocytes to a disease site. During an inflammatory response, leukocytes are recruited to the site of injury and extravasated by a series of cellular interactions involving cell-cell and cell-substrate adhesion.
  • One family of molecules that serves an important adhesive function is integrins. Integrins are expressed on cell surfaces and function in cell-cell and cell-substrate adhesion. Integrins are alpha-beta heterodimers: each integrin has an alpha (α) subunit non-covalently binded to a beta (β) subunit. When activated, integrins bind to extracellular ligands and induce adhesion (the expression of integrins on a cell surface alone is inadequate for adhesion—they must be activated to become adhesive). The integrin activation state is transient, such that there is a rapid flux between adhesive and non-adhesive states which is important for cell movement, e.g., a cell is endowed with the ability to rapidly adhere to various cell surfaces and matrices and to migrate among cells and tissue.
  • There are four known integrins having a β2 or CD18 subunit which comprise the CD11/CD18 integrin sub-family, namely, Lymphocyte Function-associated Antigen 1 (LFA-1) (CD11a/CD18 or αLβ2); Macrophage Antigen 1 (Mac-1) (CD11b/CD18 or (XMP2); p150,95 (CD11c/CD18 or (XP2); and αDβ2. The CD11/CD18 family of integrins is also referred to as Leukointegrins as they are expressed on the surface of various leukocyte cells, and they mediate a number of inflammation-related cellular interactions. See Diamond et al., “The Dynamic Regulation of Integrin Adhesiveness,” Current Biology, Vol. 4 (1994) at pp. 506-532.
  • Ligands to LFA-1 and Mac-1 comprise the intercellular adhesion molecule (ICAM) ICAM-1. The primary CD11/CD18 integrin is LFA-1, which also binds with ICAM-2 and ICAM-3. ICAMs are found on endothelium cells, leukocytes, and other cell types, and their interaction with CD11/CD18 integrins is critical to immune system function. The interaction between the CD18 integrins, particularly LFA-1, and ICAMs mediates antigen presentation, T-cell proliferation, and adhesion between the endothelium and activated leukocytes which is necessary for leukocytes to migrate from the circulatory system into tissue. A condition termed “Leukocyte Adhesion Deficiency” has been identified in patients having a deficiency in CD18 integrins. These patients are unable to mount a normal inflammatory or immune response; they suffer from disorders such as recurrent infections, poor wound healing, granulocytosis, progressive periodontitis, and umbilical cord separation. See Anderson et al., “Leukocyte LFA-1, OKMI, p150,95 Deficiency Syndrome: Functional and Biosynthesis Studies of Three Kindreds,” Fed. Proc., Vol. 44 (1985), at pp. 2671-2677.
  • While sufficient levels of CD18 integrins interacting with ICAMs are needed to mount a normal immune response, significant cellular and tissue injury can result in chronic inflammatory states where there is an inappropriate influx of leukocytes to the disease site. Continuous recruitment of leukocytes from blood vessels into inflamed tissue, as in chronic inflammatory states, can perpetuate tissue injury and lead to excessive fibrous repair and autoimmune disease. Thus, inhibition of the interaction between LFA-1 and/or Mac-1 and their ICAMs can be advantageous in treating inflammatory or immune disease. For example, monoclonal antibody blockade of either ICAM or LFA-1 has been shown to prevent the migration of leukocytes into tissue and the subsequent development of inflammatory disease in animal models of rheumatoid arthritis, inflammatory bowel disease, and pulmonary inflammation (e.g., asthma). Knockout mice deficient in ICAMs have reduced susceptibility to induced arthritis, ischemia injury, impaired lung inflammatory responses, and increased tolerance to transplantations (e.g. heart grafts). See Anderson, supra. Antibodies blocking the ICAM-LFA-1 interaction reportedly suppress cardiac allograft rejection and islet cell xenograft rejection in animal models. See Gorski, “The Role of Cell Adhesion Molecules in Immunopathology,” Immunology Today, Vol. 15 (1994), at pp. 251-255.
  • Compounds inhibiting CD18 integrins, ICAMs, and/or the LFA-1:ICAM interaction could potentially demonstrate a wide range of utilities in treating inflammatory or immune diseases. Blocking LFA-1 reportedly inhibits the influx of leukocytes in almost every system, including the skin, peritoneum, synovium, lung, kidney, and heart, and blocking ICAM-1 would be expected to have similar effects. Also, present therapies for many inflammatory and immune diseases have drawbacks. For example, current treatments for asthma include β2-agonists, inhaled corticosteroids, and LTD4 antagonists. However, β2-agonists have limited efficacy and inhaled corticosteroids raise safety concerns. To treat psoriasis, current therapies include PUVA, methotrexate, cyclosporin A, and topical treatments. The first three of these therapies raise toxicity issues over long-term (6-9 month) use, whereas topical treatments have limited efficacy. Additionally, these treatments typically are applied only in response to flares and not as a prophylaxis measure.
  • Compounds that reportedly inhibit LFA-1/ICAM for use as anti-inflammatory agents include thiadiazole-based compounds (see Intern. Pub. No. WO 99/20,618, “Thiadiazole Amides Useful as Anti-Inflammatory Agents” filed by Pharmacia & Upjohn Co.; and WO 99/20,617, also to Pharmacia and Upjohn); and thiazole compounds linked to phenyl and pyrazole rings (Sanfilippo et al., “Novel Thiazole Based Heterocyclos as Inhibitors of LFA-1/ICAM-J Mediated Cell Adhesion,” J. Med. Chem., Vol. 38 (1995) at pp.1057-1059). Small molecules that reportedly are antagonists to the binding of ICAMs with CD18 integrins include various benzylamines and 2-bromobenzoyltryptophan compounds (see Intern. Pub. No. WO99/49,856, “Antagonists for Treatment of CD11/CD18 Adhesion Receptor Mediated Disorders,” filed by Genentech, Inc.), and 1-(3,5 dichlorophenyl) imidazolidines (see Intern. Pub. No. WO98/39303, “Small Molecules Useful in the Treatment of Inflammatory Disease,” filed by Boehringer Ingelheim Pharmaceuticals, Inc. See also Boehringer patent applications WO 01/07052, WO 01/07048, WO 01/07044, WO 01/06984, and WO 01/07440). Hydantoin compounds are disclosed in WO 01/30781 A2 (published May 3, 2001) to Tanabe Seiyaku Co. Ltd, WO 03029245 A1 (published Apr. 10, 2003) to Bristol-Myers Squibb; and WO 01030781 A1 (published May 3, 2003) to Bristol-Myers Squibb.
  • As may be appreciated, those in the field of pharmaceutical research continue to seek to develop new compounds and compositions for treating inflammatory and immune disease such as inhibitors of Leukointegrins and/or ICAMs. Particularly in the area of immune response, many individuals respond differently to different drugs. Thus, there is an interest in providing consumers not only with pharmaceutical compositions demonstrating increased effectiveness and reduced side-effects but also different structures or mechanisms of action to provide consumers with a choice of options. Diazaspiroheptane compounds are disclosed in Park et al., “Preparation of a 990 Member Chemical Compound Library of Hydantoin and Isoxazoline-Containing Heterocyclos Using Multipin Technology,” J. Comb. Chem., Vol. 3(2) (2001), at pp. 171-76. Spiro heterocyclos are also disclosed in Couture et al., “Chemistry of Cyclic Aminooxycarbenes,” Can. J. Chem., Vol. 75(9) (1997) at pp. 1281-1294; Brandstetter et al., “Glucofuranose Analogs of Hydanocidin,” Tetrahedron, Vol. 52(32) (1996), at pp. 10721-10736; Brandstetter et al., “Spirohydantois of Glucofuranose: Analogs of Hydantocidin,” Tetrahedron Lett., Vol. 36(12) (1995) at pp. 2149-52; in U.S. Pat. Nos. 6,022,875, 4,241,208, 4,066,615, and 3,941,744, and International patent application WO 01/45704.
  • Each of the patents, patent applications and publications referenced above and hereinafter is incorporated herein by reference.
  • SUMMARY OF THE INVENTION
  • The present invention provides compounds useful in treating inflammatory or immune disease having the formula (I):
    Figure US20050119279A1-20050602-C00002
      • its stereoisomers, or a pharmaceutically-acceptable salt, hydrate, or prodrug thereof, in which:
      • K and L are independently O or S;
      • Q is a bond, —C(═O)— or branched or straight chain C1-6alkylene optionally substituted with one to two R4;
      • Ar is aryl or heteroaryl;
      • Y is a bond or —C(R6aR7a)—;
      • J1 is —N(R5)— or (R6bR7b)—;
      • J2 is —N(R5)— or —C(R6cR7c)—;
      • J3 is N(R5) or C(R7dR7d);
      • provided that only one of J1, J2 and J3 may be —N(R5)—, so that ring A is a five-to-six membered cycloalkyl or heterocyclo ring having from 0 to 2 heteroatoms;
      • R1 is N or C(R9);
      • R2 and R3 are independently selected from hydrogen, halogen, nitro, cyano, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —SR12, —OR12, —NR12R13, —CO2R12, —C(═O)R12, —C(═O)NR12R13, aryl, heterocyclo, cycloalkyl, and heteroaryl;
      • R4 is selected from OH, O(C1-6alkyl), halogen, cyano, CF3, OCF3, NH2, NH(6alkyl), and N(C1-6akyl)2;
      • R5 is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —NR14R15, —C(═O)R14, —CO2R14, —C(═O)NR14R15, —S(O)pR15a, —SO2NR14R15, aryl, heterocyclo, cycloalkyl, and heteroaryl; or when R5 is joined to atom J1, J2 or J3, R5 may be taken together with one of R6a, R6b R6c or R6d attached to an adjacent atom of ring A to form a fused heterocyclo or heteroaryl ring;
      • at least one of R6a with R7a, or R6b with R7b, or R6c with R7c are taken together to form a spiro-cycloalkyl or spiro-heterocyclo ring; provided that the spiro-heterocyclo ring is not C2-3 alkylenedioxy;
      • R6a, R6b, R6c, R6d, R7a, R7b, R7c, and R7d which do not form a spiro-cycloalkyl or spiro-heterocyclo ring are independently selected from hydrogen, halogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, nitro, cyano, —SR16, —OR16, —NR16R17, —C(═O)R16, —CO2R16, —C(═O)NR16R17, —NR16C(═O)R17, —NR16C(═O)OR17, —S(O)qR17a, —NR16SO2R17a, —SO2NR16R17, aryl, heterocyclo, cycloalkyl, and heteroaryl;
      • or R6a, R6b, R6c, R6d, R7a, R7b, R7c, and R7d which do not form a spiro-cycloalkyl or spiro-heterocyclo ring and which are attached to the same carbon atom may be taken together to form a keto group;
      • or R6a, R6b, R6c, R6d, R7a, R7b, R7c, and R7d which do not form a spiro-cycloalkyl or spiro-heterocyclo ring and which are attached to adjacent carbon atoms may be taken together to form a fused benzo, cycloalkyl, heterocyclo, or heteroaryl ring;
      • R9 is selected from hydrogen, halogen, nitro, cyano, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —SR18, —OR18, —NR18R19, —CO2R18, —C(═O)R18, —C(═O)NR18R19, aryl, heterocyclo, cycloalkyl, and heteroaryl;
      • R12, R13, R14, R15, R16, R17, R18, and R19 (i) are selected independently of each other from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo; or (ii) any two of R12, R13, R14, R15, R16, R17, R18, and R19 when attached to the same nitrogen atom (as in NR12R13, NR14R15, NR16R17, or NR18R19) may be taken together to form a heteroaryl or heterocyclo ring, with the remainder of R10, R11, R12, R13, R14, R15, R16, R17, R18, and R19 being selected independently from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo;
      • R15a, and R17a are independently selected from alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo;
      • p is 1, 2, or 3; and
      • q is 1, 2, or 3.
  • The present invention is also directed to pharmaceutical compositions useful in treating immune or inflammatory diseases comprising compounds of formula (I), or stereoisomers, pharmaceutically-acceptable salts, pharmaceutically-acceptable carriers or diluents thereof. The invention further relates to methods of treating immune or inflammatory diseases comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound according to formula (I).
  • DETAILED DESCRIPTION OF THE INVENTION
  • The following are definitions of terms used in the present specification. The initial definition provided for a group or term herein applies to that group or term throughout the present specification individually or as part of another group, unless otherwise indicated.
  • The terms “alkyl” and “alk” refers to a straight or branched chain alkane (hydrocarbon) radical containing from 1 to 12 carbon atoms, preferably 1 to 6 carbon atoms. Exemplary “alkyl” groups include methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, isobutyl pentyl, hexyl, isohexyl, heptyl, 4,4-dimethylpentyl, octyl, 2,2,4-trimethylpentyl, nonyl, decyl, undecyl, dodecyl, and the like. The term “C1-C6 alkyl” refers to a straight or branched chain alkane (hydrocarbon) radical containing from 1 to 6 carbon atoms, such as methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, and isobutyl..
  • “Substituted alkyl” refers to an alkyl group substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment. Exemplary substituents include but are not limited to one or more of the following groups: hydrogen, halogen (e.g., a single or multiple halo substitutents) trifluoromethyl, trifluoromethoxy, cyano, nitro, alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocyclo, aryl, ORa, SRa, S(═O)Re, S(═O)2Re, P(═O)2Re, S(═O)2ORe, P(═O)2ORe, NRbRc, NRbS(═O)2Re, NRbP(═O)2Re, S(═O)2NRbRc, P(═O)2NRbRc, C(═O)ORe, C(═O)Ra, C(═O)NRbRc, OC(═O)Ra, OC(═O)NRbRc, NRbC(═O)ORe, NRdC(═O)NRbRc, NRdS(═O)2NRbRc, NRdP(═O)2NRbRc, NRbC(═O)Ra, or NRbP(═O)2Re, wherein Ra is hydrogen, alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocyclo, or aryl; Rb, Re and Rd are independently hydrogen, alkyl, cycloalkyl, heterocyclo, aryl, or heteroaryl, or said Rb and Re together with the N to which they are bonded optionally form a heterocyclo; and Re is alkyl, cycloalkyl, alkenyl, alkynyl, heterocyclo, aryl or heteroaryl. Exemplary substituents also include spiro-attached or fused cylic substituents, especially spiro-attached cycloalkyl, spiro-attached heterocyclo, fused cycloalkyl, fused heterocyclo, fused aryl, or fused heteroaryl, In both the aforementioned groups of exemplary substitutents, groups such as alkyl, cycloalkyl, alkenyl, alkynyl, cycloalkenyl, heterocyclo, aryl and heteroaryl can themselves be optionally substituted, as described infra.
  • The term “alkenyl” refers to a straight or branched chain hydrocarbon radical containing from 2 to 12 carbon atoms and at least one carbon-carbon double bond. Exemplary such groups include ethenyl or allyl. “Substituted alkenyl” refers to an alkenyl group substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment. Exemplary substituents include, but are not limited to, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents.
  • The term “alkynyl” refers to a straight or branched chain hydrocarbon radical containing from 2 to 12 carbon atoms and at least one carbon to carbon triple bond. Exemplary such groups include ethynyl. “Substituted alkynyl” refers to an alkynyl group substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment. Exemplary substituents include, but are not limited to, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents.
  • The term “halo” or “halogen” refers to chloro, bromo, fluoro and iodo.
  • The term “haloalkyl” means a substituted alkyl having one or more halo substituents. For example, “haloalkyl” includes mono, bi, and trifluoromethyl.
  • The term “haloalkoxy” means an alkoxy group having one or more halo substituents. For example, “haloalkoxy” includes OCF3.
  • The term “cycloalkyl” refers to fully saturated and partially unsaturated hydrocarbon rings of 3 to 9, preferably 3 to 7 carbon atoms. The term “cycloalkyl” includes such rings Exemplary such groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, etc. Cycloalkyl groups may be unsubstituted or substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment. Exemplary substituents include, but are not limited to, nitro, cyano, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents. Exemplary substituents also include spiro-attached or fused cylic substituents, especially spiro-attached cycloalkyl, spiro-attached heterocyclo, fused cycloalkyl, fused cycloalkenyl, fused heterocyclo, fused aryl, or fused heteroaryl, the aforementioned cycloalkyl, heterocyclo, aryl and heteroaryl can themselves be optionally substituted.
  • The term “aryl” refers to unsubstituted or substituted cyclic, aromatic hydrocarbon groups which have 1 to 5 aromatic rings, especially monocyclic or bicyclic groups such as phenyl, biphenyl or naphthyl. Where containing two or more aromatic rings (bicyclic, etc.), the aromatic rings of the aryl group may be joined at a single point (e.g., biphenyl), or fused (e.g., naphthyl, phenanthrenyl and the like). The aryl group may be substituted by one or more substituents, preferably 1 to 3 substituents, at any point of attachment. Exemplary substituents include, but are not limited to, nitro, cycloalkyl, cyano, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents. Exemplary substituents also include fused cylic, especially fused cycloalkyl, fused heterocyclo, fused aryl, or fused heteroaryl.
  • The term “heterocyclo” refers non-aromatic 3 to 7 membered monocyclic groups, 7 to 11 membered bicyclic groups, and 10 to 15 membered tricyclic groups, in which at least one of the rings has at least one heteroatom (O, S or N). Each heerocyclo group may be unsubstituted or substituted. Each ring of the heterocyclo group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less, and further provided that the ring contains at least one carbon atom. The fused rings completing bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated. The nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quaternized. The heterocyclo group may be attached at any available nitrogen or carbon atom in any fashion, including spiro- attachments. The heterocyclo group may be substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment. Exemplary substituents include, but are not limited to, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, nitro, oxo (i.e., ═O), cyano, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents. Exemplary substituents also include spiro-attached or fused cylic substituents at any available point or points of attachment, especially spiro-attached cycloalkyl, spiro-attached cycloalkenyl, spiro-attached heterocyclo, fused cycloalkyl, fused cycloalkenyl, fused heterocyclo, fused aryl, or fused heteroaryl, the aforementioned cycloalkyl, cycloalkenyl, heterocyclo, aryl and heteroaryl can themselves be optionally substituted.
  • Exemplary monocyclic heterocyclo groups include azetidinyl, pyrrolidinyl, oxetanyl, imidazolinyl, oxazolidinyl, isoxazolinyl, thiazolidinyl, isothiazolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolodinyl, 2-oxoazepinyl, azepinyl, 4-piperidonyl, tetrahydropyranyl, morpholinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, 1,3-dioxolane and tetrahydro-1,1-dioxothienyl and the like. Exemplary bicyclic heterocyclo groups include quinuclidinyl.
  • The term “heteroaryl” refers to substituted and unsubstituted aromatic 5 or 6 membered monocyclic groups, 9 or 10 membered bicyclic groups, and 11 to 14 membered tricyclic groups which have at least one heteroatom (O, S or N) in at least one of the rings. Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom. The fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated. The nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quatemized. Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non-aromatic. The heteroaryl group may be attached at any available nitrogen or carbon atom of any ring. “Substituted heteroaryl” refers to heteroaryl groups substituted with one or more substituents, preferably 1 to 4 substituents, at any available point of attachment. Exemplary substituents include, but are not limited to, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, nitro, cyano, alkyl or substituted alkyl, as well as those groups recited above as exemplary alkyl substituents. Exemplary substituents also include or fused cylic substituents at any available points of attachment, especially fused cycloalkyl, fused cycloalkenyl, fused heterocyclo, fused aryl and fused heteroaryl, the aforementioned cycloalkyl, cycloalkenyl, heterocyclo, aryl and heteroaryl can themselves be optionally substituted.
  • Exemplary monocyclic heteroaryl groups include pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl and the like.
  • Exemplary bicyclic heteroaryl groups include indolyl, benzothiazolyl, benzodioxolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl, dihydroisoindolyl, tetrahydroquinolinyl and the like.
  • Exemplary tricyclic heteroaryl groups include carbazolyl, benzidolyl, phenanthrollinyl, acridinyl, phenanthridinyl, xanthenyl and the like.
  • Unless otherwise indicated, when reference is made to a specifically-named aryl (e.g., phenyl), cycloalkyl (e.g., cyclohexyl), heterocyclo (e.g., pyrrolidinyl) or heteroaryl (e.g., imidazolyl), unless otherwise specifically indicated the reference is intended to include rings having 0 to 3, preferably 0-2, substituents selected from those recited above for the aryl, cycloalkyl, heterocyclo and/or heteroaryl groups, as appropriate.
  • The term “heteroatoms” shall include oxygen, sulfur and nitrogen.
  • The term “carbocyclic” means a saturated or unsaturated monocyclic or bicyclic ring in which all atoms of all rings are carbon. Thus, the term includes cycloalkyl and aryl rings. The carbocyclic ring may be substituted in which case the substituents are selected from those recited above for cycloalkyl and aryl groups.
  • When the term “unsaturated” is used herein to refer to a ring or group, the ring or group may be fully unsaturated or partially unsaturated.
  • When a functional group is termed “protected”, this means that the group is chemically modified to mitigate, especially preclude, undesired side reactions at the protected site. Suitable protecting groups for the methods and compounds described herein include, without limitation, those described in standard textbooks, such as Greene, T. W. et al., Protective Groups in Organic Synthesis, Wiley, N.Y. (1999).
  • Unless otherwise indicated, any heteroatom with unsatisfied valences is assumed to have hydrogen atoms sufficient to satisfy the valences.
  • Throughout the specification, groups and substituents thereof may be chosen by one skilled in the field to provide stable moieties and compounds and compounds useful as pharmaceutically-acceptable compounds and/or intermediate compounds useful in making pharmaceutically-acceptable compounds.
  • The compounds of formula I form salts which are also within the scope of this invention. Reference to a compound of the formula I herein is understood to include reference to salts thereof, unless otherwise indicated. The term “salt(s)”, as employed herein, denotes acidic and/or basic salts formed with inorganic and/or organic acids and bases. In addition, when a compound of formula I contains both a basic moiety, such as but not limited to a pyridine or imidazole, and an acidic moiety such as but not limited to a carboxylic acid, zwitterions (“inner salts”) may be formed and are included within the term “salt(s)” as used herein. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred, although other salts are also useful, e.g., in isolation or purification steps which may be employed during preparation. Salts of the compounds of the formula I may be formed, for example, by reacting a compound I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
  • The compounds of formula I which contain a basic moiety, such as but not limited to an amine or a pyridine or imidazole ring, may form salts with a variety of organic and inorganic acids. Exemplary acid addition salts include acetates (such as those formed with acetic acid or trihaloacetic acid, for example, trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, hydroxyethanesulfonates (e.g., 2-hydroxyethanesulfonates), lactates, maleates, methanesulfonates, naphthalenesulfonates (e.g., 2-naphthalenesulfonates), nicotinates, nitrates, oxalates, pectinates, persulfates, phenylpropionates (e.g., 3-phenylpropionates), phosphates, picrates, pivalates, propionates, salicylates, succinates, sulfates (such as those formed with sulfuric acid), sulfonates (such as those mentioned herein), tartrates, thiocyanates, toluenesulfonates such as tosylates, undecanoates, and the like.
  • The compounds of formula I which contain an acidic moiety, such but not limited to a carboxylic acid, may form salts with a variety of organic and inorganic bases. Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as benzathines, dicyclohexylamines, hydrabamines (formed with N,N-bis(dehydroabietyl) ethylenediamine), N-methyl-D-glucamines, N-methyl-D-glycamides, t-butyl amines, and salts with amino acids such as arginine, lysine and the like. Basic nitrogen-containing groups may be quaternized with agents such as lower alkyl halides (e.g. methyl, ethyl, propyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g. dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (e.g. decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides), aralkyl halides (e.g. benzyl and phenethyl bromides), and others.
  • Prodrugs and solvates of the compounds of the invention are also contemplated herein. The term “prodrug” as employed herein denotes a compound which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of the formula I, or a salt and/or solvate thereof. Solvates of the compounds of formula I include, for example, hydrates.
  • Compounds of the formula I, and salts thereof, may exist in their tautomeric form (for example, as an amide or imino ether). All such tautomeric forms are contemplated herein as part of the present invention.
  • All stereoisomers of the present compounds (for example, those which may exist due to asymmetric carbons on various substituents), including enantiomeric forms and diastereomeric forms (i.e. “stereoisomers”), are contemplated within the scope of this invention. Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers (e.g., as a pure or substantially pure optical isomer having a specified activity), or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The chiral centers of the present invention may have the S or R configuration as defined by the IUPAC 1974 Recommendations. The racemic forms can be resolved by physical methods, such as, for example, fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography. The individual optical isomers can be obtained from the racemates by any suitable method, including without limitation, conventional methods, such as, for example, salt formation with an optically active acid followed by crystallization.
  • All configurational isomers of the compounds of the present invention are contemplated, either in admixture or in pure or substantially pure form. The definition of compounds of the present invention embraces both cis (Z) and trans (E) alkene isomers, as well as cis and trans isomers of cyclic hydrocarbon or heterocyclo rings.
  • Throughout the specifications, groups and substituents thereof may be chosen to provide stable moieties and compounds.
  • Preferred Compounds
  • Preferred compounds of the invention include compounds of formula (I) in which one or more, preferably all, of the following substituents are as defined below:
      • K iand L are both O;
      • Q is a straight chain C1-6alkylene (preferably methylene);
      • Ar is phenyl optionally substituted one to three R20;
      • R2 and R3 are selected from halogen, (C1-6)alkyl, cyano, halo(C1-6)alkyl, halo(C1-6)alkoxy, nitro, phenyloxy, benzyloxy, and phenylthio;
      • R20 at each occurrence is independently selected from halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, —C(═O) (C1-6)alkyl, —C(═O)NH(CH2)rCO2H, —C(═O)NH(CH2)rCO2(C1-6alkyl), and S(O)2(C1-6alkyl); or from phenyl, benzyl, phenyloxy, benzyloxy and heteroaryl in turn optionally substituted with one to two of halogen, (C1-6)alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, and/or —C(═O) (C1-6)alkyl; or alternatively, two R20 groups join together with each other to form a fused benzo ring; and
      • r is 1, 2, 3, or4.
  • Particularly preferred compounds, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, within the scope of formula (I) are those wherein Q-Ar together form:
    Figure US20050119279A1-20050602-C00003
      • wherein
      • R20a and R20b are independently selected from halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, —C(═O) (C1-6)alkyl, C(═O)NH(CH2)rCO2H, —C(═O)NH(CH2)rCO2(C1-6alkyl), and S(O)2(C1-6alkyl); or from phenyl, benzyl, phenyloxy, benzyloxy and heteroaryl in turn optionally substituted with one to two of halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —NH2, —NH(C1-6alkyl), —N(C1-6alkyl)2, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, and/or —C(═O) (C1-6)alkyl; or alternatively, two R20b groups join together with each other or one R20b joins together with R20a to form a fused benzo ring;
      • n is 0, 1, or 2; and
      • r is 1, 2, 3, or 4.
  • Other particularly preferred compounds, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, within thie scope of formula (I) are those wherein one or more, preferably all, of the following substituents are defined as below:
      • Y is a bond;
      • J1 is —CHR6b—;
      • J2 is is —C(R6cR7c)-wherein R6c with R7c are taken together to form a spirocycloalkyl or spiroheterocyclo ring;
      • J3 is CH(R6d);
      • R1 is C(R8) (preferably CH);
      • R2 and R3 are independently halogen.
      • R6b, and R6d are independently selected from
        • a) hydrogen, halogen, and cyano;
        • b) —SR16, —OR16, —NR16R17, —C(═O)R16, —CO2R16, —C(═O)NR16R17, —NR16C(═O)R17, —NR16C(═O)OR17, —S(O)qR17a, —NR16SO2R17a, and —SO2NR16R17; and
        • c) (C1-6)alkyl, phenyl, four to seven membered heterocyclo, C3-7cycloalkyl, and five to six membered heteroaryl, each of which in turn is optionally substituted with one to two groups selected from R22;
      • R16 and R17 are selected independently from hydrogen, C1-6alkyl, phenyl, four to seven membered heterocyclo, C3-7cycloalkyl, and five to six membered heteroaryl, each of which in turn is optionally substituted with one to two groups selected from R23;
      • R17a is C1-6alkyl, phenyl, four to seven membered heterocyclo, C3-7cycloalkyl, five to six membered heteroaryl each of which is optionally substituted with one to two groups selected from R23; and
      • R22 and R23 are at each occurrence selected independently from halogen, cyano, C1-6alkyl, hydroxy, trifluoromethyl, trifluoromethoxy, —O(C1-6alkyl), —C(═O)H, —C(═O)(C1-6alkyl), —CO2H, —CO2(C1-6alkyl), —C(═O)NH2, —C(═O)NH2, —C(═O)NH(C1-6alkyl), —C(═O)N(C1-6alkyl)2, —NH2, —NH(C1-6alkyl), —N(C1-6alkyl)2, hydroxy(C1-6)alkyl, methoxy(C1-6)alkyl, ethoxy(C1-6)alkyl, amino(C1-6)alkyl, and halo(C1-6)alkyl.
  • Alternatively, preferred compounds of the invention include compounds of forula (Ia)
    Figure US20050119279A1-20050602-C00004
      • or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, in which
      • K and L are independently O or S;
      • Q is a bond, —C(═O)— or branched or straight chain C1-6alkylene optionally substituted with one to two R4 (Q is preferably C1-6alkylene, especially methylene);
      • Ar is aryl or heteroaryl;
      • Y is a bond or —C(R6aR7a)—;
      • J1 is —N(R5)— or —C(R6bR7b)—;
      • J3 is —N(R5)— or —C(R6dR7d)—;provided, that only one of J1 and J3 may be —N(R5)—, so that ring A is a five-to-six membered cycloalkyl or heterocyclo ring having from 0 to 2 heteroatoms;
      • Z1 and Z2 are independently NR25, S or O; so that Z forms a spiro heterocyclic ring provided that Z is not a C2-3 alkylenedioxy ring and that only one of Z1 and Z2 may be NR25;
      • R1 is N or C(R9);
      • R2 and R3 are independently selected from hydrogen, halogen, nitro, cyano, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —SR12, —OR12, —NR12R13, —CO2R12, —C(═O)R12, —C(═O)NR12R13, aryl, heterocyclo, cycloalkyl, and heteroaryl;
      • R4 is selected from OH, O(C1-6alkyl), halogen, cyano, CF3, OCF3, NH2, NH(C1-6alkyl), and N(C1-6alkyl)2;
      • R5 is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —NR14R15, —C(═O)R14, —CO2R14, —C(═O)NR14R15, —S(O)pR15a, —SO2NR14R15, aryl, heterocyclo, cycloalkyl, and heteroaryl; or when R5 is joined to atom J1, J3 or Y, R5 may be taken together with one of R6a, R6b or R6d attached to an adjacent atom of ring A to form a fused heterocyclo or heteroaryl ring;
      • R6a, R6b, R6d, R7a, R7b, R7b and R7d are independently selected from hydrogen, halogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, nitro, cyano, —SR16, —OR16, —NR16R17, —C(═O)R16, —CO2R16, —C(═O)NR16R17, —NR16C(═O)R17, —NR16C(═O)OR17, —S(O)qR17a, —NR16SO2R17a, —SO2NR16R17, aryl, heterocyclo, cycloalkyl, and heteroaryl;
      • R9 is selected from hydrogen, halogen, nitro, cyano, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —SR18, —OR18, —NR18R19, —CO2R18, —C(═O)R18, —C(═O)NR18R19, aryl, heterocyclo, cycloalkyl, and heteroaryl;
      • R12, R13, R14, R15, R16, R17, R18, and R19 (i) are selected independently of each other from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo; or (ii) any two of R12, R13, R14, R15, R16, R17, R18, and R19 when attached to the same nitrogen atom (as in NR12R13, NR14R15, NR16R17, or NR18R19) may be taken together to form a heteroaryl or heterocyclo ring, with the remainder of R12, R13, R14, R15, R16, R17, R18, and R19 being selected independently from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo;
      • R11a, R15a, and R17a are independently selected from alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo;
      • R25 is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —NR26R27, —C(═O)R26, —CO2R26, —C(═O)NR26R27, —S(O)pR27, —SO2NR26R27, aryl, heterocyclo, cycloalkyl, and heteroaryl;
      • R26, and R27 are selected independently of each other front hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo;
      • p is 0, 1, 2, or 3; and
      • q is 1, 2, or 3.
  • Particularly preferred compounds within the scope of formula (Ia), or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, are those in which one or more, preferably all, of the following substituents re as defined below:
      • Y is a bond; and
      • Q-Ar together form:
        Figure US20050119279A1-20050602-C00005
      • wherein
      • R1 is CH.
      • R2 and R3 are independently halogen;
      • J1 is —C(R6bR7b)—;
      • J3 is —C(R6bR7b)—;
      • one of Z1 or Z2 is NR25;
      • R20a and R20b are independently selected from halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, —C(═O) (C1-6)alkyl, —C(═O)NH(CH2)rCO2H, —C(═O)NH(CH2)rCO2(C1-6alkyl), and S(O)2(C1-6alkyl); or from phenyl, benzyl, phenyloxy, benzyloxy and heteroaryl in turn optionally substituted with one to two of halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —NH2, —NH(C1-6alkyl), —N(C1-6alkyl)2, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, and/or —C(═O) (C1-6)alkyl; or alternatively, two R20b groups join together with each other or one R20b joins together with R20a to form a fused benzo ring;
      • n is 0, 1, or 2; and
      • r is 1, 2, 3, or 4.
  • Alternatively, preferred compounds of the invention include compounds of formula (Ib)
    Figure US20050119279A1-20050602-C00006
      • its stereoisomers, or a pharmaceutically-acceptable salt, hydrate, solvate, or prodrug thereof, in which:
      • one of Z1 or Z2 is NR25 and the other of Z1 or Z2 is O or S;
      • R1 is N or C(R9);
      • R2 and R3 are independently selected from halogen, (C1-6)alkyl, cyano, halo(C1-6)alkyl, halo(C1-6)alkoxy, nitro, phenyloxy, benzyloxy, and phenylthio;
      • R6 is selected from (a) halogen, nitro, and cyano; or from (b) —SR16, —OR16, —NR16R17, —C(═O)R16, —CO2R16, —C(═O)NR16R17, —NR16C(═O)R17, —NR16C(═O)OR17, —S(O)qR17a, —NR16SO2R17a, and —SO2NR16R17; or from (c) alkyl, alkenyl, aryl, heterocyclo, cycloalkyl, and heteroaryl, in turn optionally substituted with one to two groups selected from R22; and/or (d) two R6 groups taken together form keto (═O), with the remainder of the R6 groups selected from (a), (b), and (c);
      • R16 and R17 are selected independently of each other from hydrogen, C1-6alkyl, phenyl, four to seven membered heterocyclo, C3-7cycloalkyl, and five to six membered heteroaryl, each of which in turn is optionally substituted with one to two groups selected from R23;
      • R17a is C1-6alkyl, phenyl, four to seven membered heterocyclo, C3-7cycloalkyl, five to six membered heteroaryl each of which is optionally substituted with one to two groups selected from R23;
      • R20a and R20b are independently selected from halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, —C(═O) (C1-6)alkyl, —C(═O)NH(CH2)rCO2H, —C(═O)NH(CH2)rCO2(C1-6alkyl), and S(O)2(C1-6alkyl); or from phenyl, benzyl, phenyloxy, benzyloxy and heteroaryl in turn optionally substituted with one to two of halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —NH2, —NH(C1-6alkyl), —N(C1-6alkyl)2, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, and/or —C(═O) (C1-6)alkyl; or alternatively, two R20b groups join together with each other or one R20b joins together with R20a to form a fused benzo ring;
      • R22, R23 and R24 are at each occurrence selected independently from halogen, cyano, nitro, C1-6alkyl, hydroxy, trifluoromethyl, trifluoromethoxy, —O(C1-6alkyl), —C(═O)H, —C(═O)(C1-6alkyl), —CO2H, —CO2(C1-6alkyl), —C(═O)NH2, —C(═O)NH2,—C(═O)NH(C1-6alkyl), —C(═O)N(C1-6alkyl)2, —NH2, —NH(C1-6alkyl), —N(C1-6alkyl)2, hydroxy(C1-6)alkyl, methoxy(C1-6)alkyl, ethoxy(C1-6)alkyl, amino(C1-6)alkyl, and halo(C1-6)alkyl;
      • R25 is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —NR26R27, —C(═O)R26, —CO2R26, —C(═O)NR26R27, —S(O)pR27, —SO2NR26R27, aryl, heterocyclo, cycloalkyl, and heteroaryl;
      • R26 and R27 are selected independently of each other from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo
      • n, s and and p are independently 0, 1, or 2;
      • q is 1, 2, or 3; and
      • r is 0, 1, 2, 3 or 4.
  • Particularly preferred compounds within the scope of formula (Ib), or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, are those in which one or more, preferably all, of the following substituents are as defined below:
      • R1 is CH;
      • R2 and R3 are independently halogen;
      • R6 is hydrogen, C1-6alkyl, substituted C1-6alkyl, —C(═O)R26 or CO2R26;
      • R20a is cyano, halogen, aryl, or heteroaryl.
      • R25 is hydrogen, C1-6alkyl, substituted C1-6alkyl, or —C(═O)R26; and
      • n is 0.
  • Especially preferred compounds are those having the formula, (i) (2R,5S,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
      • (2R,5R,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
      • (2R,5R,7a′S)-7′a-[(4-Bromophenyl)methyl]-2′-(3,5-dichlorophenyl)dihydro-5-methylspiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
      • (2S ,5R,7a′S)-7′a-[(4-Bromophenyl)methyl]-2′-(3,5-dichlorophenyl)dihydro-5-methylspiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
      • (2R,7a′S)-7′a-[(4-Bromophenyl)methyl]-2′-(3,5-dichlorophenyl)dihydrospiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
      • (2S ,7a′S)-7′a-[(4-Bromophenyl)methyl]-2′-(3,5-dichlorophenyl)dihydrospiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
      • 4-[[(2R,5R,7a′S)-2′-(3,5-Dichlorophenyl)dihydro-5-methyl-1′,3′,4-trioxospiro[oxazolidine-2,6′(5′H)-[[H]pyrrolo[1,2-c]imidazol]-7′a(7′H)-yl]methyl]benzonitrile;
      • 4-[[(2S ,5R,7a′S)-2′-(3,5-Dichlorophenyl)dihydro-5-methyl-1′,3′,4-trioxospiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo[1,2-c]imidazol]-7′a(7′H)-yl]methyl]-benzonitrile;
      • (2R,5R,7a′S)-2′-(3,5-dichlorophenyl)-7a′-[[4-(pyrimidin-4-yl)-phenyl)-methyl]-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
      • ethyl (4′S,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate;
      • ethyl (4′S,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro [pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate;
      • ethyl (4′R,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate;
      • ethyl (4′R,6S,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate;
      • (±) ethyl (4′S,7aS)-3′-acetyl-7a-ethyl-2-methyl-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate;
      • (±) ethyl (4′S,7aS)-7a-(4-bromophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate; or
      • (ii) a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof.
        Methods of Preparation
  • The compounds of the present invention may be prepared by methods such as those illustrated in the following schemes. Solvents, temperatures, pressures, and other reaction conditions may readily be selected by one of ordinary skill in the art. Starting materials are commercially available or readily prepared by one of ordinary skill in the art. The symbols used in the followoing schemes are defined as above, unless otherwise indicated.
    Figure US20050119279A1-20050602-C00007

    Compounds of the general formula (Ia) may be obtained from compounds of formula (1) by dehydrative cyclization. The choice of the dehydrating agent will be readily apparent to one skilled in the art of organic chemistry. Such methods are, for example described by Greene, Theodora W. and Wuts, Peter G. M. in “Protective groups in organic synthesis” 3rd Ed., (1999) Publisher (John Wiley and Sons, Inc., New York, N.Y). For example, if the reagent (2) is an amino-alcohol (Z1H=NH2, Z2H=OH), compounds of formula (Ia) may be obtained by refluxing (1) and (2) in a solvent such as xylene, a catalytic amount of acid such a para-toluenesulfonic acid and at a temperature range of 100-140° C. with the azeotropic removal of water. Furthermore, if the reagent is an amino-thiol (Z1H=NH2, Z2H=SH), compounds of formula (Ia) may be obtained for example by stirring (1) and (2) in a solvent such as ethanol at a temperature range of 20-50° C. (Refouvelet, Bernard et al.: Chem.Pharm.Bull., 1994, 42 (5), 1076-1083)
  • Compounds of the general formula (1) may be obtained by the oxidation of compounds of general formula (3) as shown in Scheme 2.
    Figure US20050119279A1-20050602-C00008

    The choice of the oxidizing agent will be readily apparent to one skilled in the art of organic chemistry. Such methods are, for example described by Hudlicky, M., in “oxidations in organic chemistry”, ACS monograph 186 (1990), American Chemical Society, Washington, D.C. For example, compounds of formula (1) may be obtained from compounds of formula (3) by using an oxidizing agent such as pyridinium chlorochromate, in a solvent like dichloromethane at temperatures ranging from 0° C. to 40° C.
  • Compounds of general formula (3) may be obtained by the protocol outlined in Scheme 3.
    Figure US20050119279A1-20050602-C00009
  • Compounds of formula (3) can be prepared by reacting the proline derivative (7) with an isocyanate (R1NCO) in DMF in the presence of K2CO3. See Issartel et al., Eur. J. Med. Chem. Chim. Ther., Vol. 31; 9 (1996), at pp. 717-724, incorporated herein by reference. The isocyantes useful for the preparation of compounds to this invention may be commercially available or readily prepared by many methods known to one skilled in the art of organic chemistry.
  • Compounds of formula (7) can be prepared by heating the oxazolidinone of formula (6) in refluxing methanolic HCl. See Remuzon et al., J. Med. Chem., Vol. 35(15) (1992) p. 2898, incorporated herein by reference.
  • Compounds of formula (6) can be prepared from compounds of formula (5) by alkylating without loss of the optical activity the oxazolidinone of formula (5) with BrCH2—Y in the presence of LDA in THF. See Wang et al., Synlett, Vol. 1 (1999), pp. 33-36, incorporated herein by reference.
  • Compounds of formula (5) can be prepared from compounds of formula (4) by cyclizing the proline compound of formula (4) with anhydrous chloral in acetonitrile. See Orsini et al., J. Heterocyclic Chem., (1989), pp. 837-841, incorporated herein by reference.
  • Compounds of formula (4) can be obtained from commercially available sources or can readily be prepared by one skilled in the field.
  • Compounds of formula (4) can be obtained in a similar way as described above in Scheme 3 from 7 by replacing the isocyanate in Scheme 3 with an isothiocyanate (Scheme 4). Compounds of formula (X) can be processed as outlined in Schemes 1 and 2 above to give the thiohydantoin analog of compound 1a
    Figure US20050119279A1-20050602-C00010
  • Alternatively compounds of formula (3) may be prepared by the sequence outlined in Scheme 5.
    Figure US20050119279A1-20050602-C00011
  • Compounds of formula (3) can be prepared from compounds of formula (9) by reacting (9) with BrCH2—R4 in the presence of LiHMDS or LDA and THF. See Kobayashi et al., Bull. Chem. Soc. Jpn., Vol. 67; 11 (1994), at pp. 3082-3087, incorporated herein by reference. The protecting group on the oxygen can then be deprotected by one skilled in the art of organic chemistry. Such methods are, for example described by Greene, Theodora W. and Wuts, Peter G. M. in “Protective groups in organic synthesis” 3rd Ed., (1999) Publisher (John Wiley and Sons, Inc., New York, N.Y).
  • Compounds of formula (9) can be prepared from compounds of formula (8) by reacting proline derivatives (8) with isocyanate R2NCO in an aqueous basic media, then cyclizing the intermediate ureidoacid in refluxing toluene in the presence of a catalytic amount of sulfuric acid. See French patent No. FR230083 and Great Britain Pat. No. B 1 503 244, titled “1,5-Alkylene-3-aryl Hydantoin Derivatives,” both to Mitsubishi Chemical Indus., Inc., incorporated herein.
  • Compounds of formula (8) can be obtained from commercially available sources or can readily be prepared by one skilled in the field.
  • Compounds of formula (Y) can be obtained from (9) by reaction with either Lawesson's reagent or phosphorus pentasulfide (Scheme 6). Compounds of formula (X) can be processed as outlined in Schemes 1 and 2 above to give the dithiohydantoin analog of compound 1a
    Figure US20050119279A1-20050602-C00012
  • Utility
  • The compounds and compositions of this invention are antagonists of LFA-1, Mac-1, and/or ICAMs. Thus, they are useful in treating various inflammatory diseases and disorders associated with the action of LFA-1, Mac-1, and/or ICAMs, particularly LFA-1:ICAM-1. The term “Leukointegrin/ICAM-associated condition” is used herein for ease of reference to refer to those diseases or disorders that are associated with the action or levels of LFA-1, Mac-1 and/or ICAM-1, ICAM-2, or ICAM-3. As used herein the term “treating” includes prophylactic and therapeutic uses and refers to the alleviation of symptoms of a particular disorder in a patient, the improvement of an ascertainable measurement associated with a particular disorder, or the prevention of a particular immune response (such as transplant rejection). The term “patient” refers to a mammal, preferably a human.
  • The inventive compounds and compositions are useful for treating a wide range of conditions, as the action of LFA-1 and/or ICAMs is associated with the influx of leukocytes in almost every system, including the skin, peritoneum, synovium, lung, kidney and heart. The inventive compounds may be used to treat conditions resulting from a response of the specific immune system in a patient or the nonspecific immune system. Such conditions include, for example, graft vs host reactions and transplant rejection (e.g., kidney, liver, heart, lung, pancreas, bone marrow, cornea, small bowel, skin allografts, skin homografts and heterografts, etc.); psoriasis, organ-tissue autoimmune diseases (e.g., Raynaud's syndrome), autoimmune thyroiditis, dermatitis, multiple sclerosis, rheumatoid arthritis, insulin dependent diabetes mellitus, uveitis, inflammatory bowel disease including Crohn's disease and ulcerative colitis and systemic lupus erythematosus, adult respiratory distress syndrome, shock, oxygen toxicity, multiple organ injury syndrome secondary to septicemia, multiple organ injury syndrome secondary to trauma, reperfusion injury of tissue due to cardiopulmonary bypass, myocardial infraction or use with thrombolysis agents, acute glomerulonephritis, vasculitis, reactive arthritis, dermatosis with acute inflammatory components, stroke, thermal injury, hemodialysis, leukapheresis, ulcerative colitis, necrotizing enterocolitis and granulocyte transfusion associated syndrome, pulmonary fibrosis, atherosclerosis, meningitis, encephalitis, experimental autoimmune encephalomyelitis, Sjorgen's syndrome, and juvenile onset diabetes. The compounds of the present invention also may be used to treat allergic conditions such as eczema and asthma or as an adjunct to minimize toxicity with cytokine therapy in the treatment of cancers. In view of their inhibition activity, the compounds may be used to treat inflammatory conditions that involve the infiltration of T-cells and chronic inflammatory responses, hypersensitivity reactions, such as skin hypersensitivity reactions (including poison ivy and poison oak), immune responses associated with delayed hypersensitivity mediated by cytokines and T-lymphocytes, and metastases.
  • The compounds of this invention further have utility in treating hypogonadism, frailty, osteoporosis, sexual dysfunction, wasting, such as wasting syndromes associated with cancer and AIDS, and anemia. The compounds further have utility in treating cancers, including but not limited to cancers of the breast, brain, skin, ovary, endometrium, bladder, prostate, lung, colon, lymphatic system, liver and kidney. The inventive compounds are useful for conditions such as hirsutism, Alzheimer's disease, non-insulin dependent diabetes mellitus, acne, seborrhea, alopecia, fibroids, hyperpilosity, cachexia, polycystic ovarian syndrome, anorexia, contraception, drug withdrawal syndrome, pregnancy termination, and benign prostate hypertrophy. The compounds are further useful as antiangiogenic agents. Additionally, the compounds may be useful as inhibitors of protein prenyltransferases, particularly famesyltransferase and the prenylation of the oncogene protein Ras. As such, the inventive compounds may potentially be useful for treating and/or preventing the diseases and disorders referred to in WO 01/45704, which is incorporated herein by reference.
  • When used as anti-inflammatory agents, the compounds may be administered prior to the onset of, at, or after the initiation of inflammation. When used prophylactically, the compounds are preferably provided in advance of any inflammatory response or symptom (for example, prior to, at, or shortly after the time of an organ or tissue transplant but in advance of any symptoms or organ rejection). Administration of the compounds may prevent or attenuate inflammatory responses or symptoms.
  • The present invention thus provides methods for treating such conditions as those listed above, comprising administering to a subject in need thereof an effective amount of at least one compound of formula (I) or a salt thereof. Other therapeutic agents such as those described below may be employed in combination with the compounds of formula (I). In the methods of the present invention, such other therapeutic agent(s) may be administered prior to, simultaneously with, or following the administration of the inventive compounds.
  • The present invention also provides pharmaceutical compositions capable of treating the Leukointegrin/ICAM-associated conditions and above-described diseases and disorders. The inventive compositions may contain other therapeutic agents and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulation.
  • The compounds of formula (I) may be administered by any means suitable for the condition to be treated, which may depend on the need for site-specific treatment or quantity of drug to be delivered. Topical administration is generally preferred for skin-related diseases, and systematic treatment preferred for cancerous or pre-cancerous conditions, although other modes of delivery are contemplated. For example, the compounds may be delivered orally, such as in the form of tablets, capsules, granules, powders, or liquid formulations including syrups; topically, such as in the form of solutions, suspensions, gels or ointments; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrastemal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; rectally such as in the form of suppositories; or liposomally. Dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents may be administered. The compounds may be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved with suitable pharmaceutical compositions or, particularly in the case of extended release, with devices such as subcutaneous implants or osmotic pumps.
  • Exemplary compositions for topical administration include a topical carrier such as PLASTIBASE® (mineral oil gelled with polyethylene).
  • Exemplary compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art. The inventive compounds may also be orally delivered by sublingual and/or buccal administration, e.g., with molded, compressed, or freeze-dried tablets. Exemplary compositions may include fast-dissolving diluents such as mannitol, lactose, sucrose, and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (AVICEL®) or polyethylene glycols (PEG); an excipient to aid mucosal adhesion such as hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), sodium carboxymethyl cellulose (SCMC), and/or maleic anhydride copolymer (e.g., GANTREZ®); and agents to control release such as polyacrylic copolymer (e.g., CARBOPOL 934®). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • Exemplary compositions for nasal aerosol or inhalation administration include solutions which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance absorption and/or bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
  • Exemplary compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • Exemplary compositions for rectal administration include suppositories which may contain, for example, suitable non-irritating excipients, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures but liquefy and/or dissolve in the rectal cavity to release the drug.
  • The effective amount of a compound of the present invention may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for a patient of from about 0.05 to 100 mg/kg of body weight of active compound per day, which may be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day. It will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors, including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and the particular condition sought to be treated and its severity. Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats, horses, and the like, subject to Leukointegrin/ICAM associated conditions and/or subject to any of the above-referenced diseases and disorders.
  • The inventive compounds and compositions may be employed alone or in combination with each other and/or other suitable therapeutic agents useful in treating Leukointegrin/ICAM-associated conditions and diseases and disorders referenced above. Exemplary of such other therapeutic agents include corticosteroids, cyclosporin, methotrexate; CELLCEPT™ (mycophenolate mofetil), co-stimulation blockades, growth hormones, and growth hormone secretagogues. Additionally, the inventive compounds may be administered either alone or in combination with anti-cancer and cytotoxic agents and treatments useful in treating cancer or other proliferative diseases, for example, where the second drug has the same or different mechanism of action than the present compounds. Examples of classes of anti-cancer and cytotoxic agents useful in combination with the present compounds include but are not limited to: alkylating agents such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; antimetabolites such as folate antagonists, purine analogues, and pyrimidine analogues; antibiotics such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; enzymes such as L-asparaginase; famesyl-protein transferase inhibitors; hormonal agents such as glucocorticoids, estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, octreotide acetate; microtubule-disruptor agents, such as ecteinascidins or their analogs and derivatives; microtubule-stabilizing agents such as taxanes, for example, paclitaxel (Taxol®), docetaxel (Taxotere®), and their analogs, and epothilones, such as epothilones A-F and their analogs; plant-derived products, such as vinca alkaloids and epipodophyllotoxins; topiosomerase inhibitors; prenyl-protein transferase inhibitors; and miscellaneous agents such as hydroxyurea, procarbazine, mitotane, hexamethylmelamine, platinum coordination complexes such as cisplatin and carboplatin; and other agents used as anti-cancer and cytotoxic agents such as biological response modifiers, growth factors; immune modulators and monoclonal antibodies. The compounds of the invention may be used in conjunction with radiation therapy.
  • Representative examples of these classes of anti-cancer and cytotoxic agents that may be used in combination with the inventive compounds include but are not limited to paclitaxel, cisplatin, carboplatin, doxorubicin, carmninomycin, daunorubicin, idarubicin, aminopterin, methotrexate, methopterin, mitomycin C, ecteinascidin 743, porfiromycin, 5-fluorouracil, 6-mercaptopurine, gemcitabine, cytosine arabinoside, podophyllotoxin or podophyllotoxin derivatives such as etoposide, etoposide phosphate or teniposide, melphalan, vinblastine, vincristine, leurosidine, vindesine and leurosine, mechlorethamine hydrochloride, cyclophosphamhide, chlorambucil, ifosfamide, busulfan, carmustin, lomustine, semustine, streptozocin, thiotepa, dacarbazine, methotrexate, thioguanine, , fludarabine, pentastatin, cladribin, cytarabine, bleomycin, mitomycin C, actinomycin D, safracins, saframycins, quinocarcins, discodermolides, vinorelbine, tamoxifen, estramustine, flutamide, buserelin, leuprolide, pteridines, diynes, levamisole, aflacon, interferon, interleukins, aldesleukin, filgrastim, sargramostim, rituximab, BCG, tretinoin, irinotecan, betamethasone, altretamine, and topotecan and any analogs or derivatives thereof.
  • Examples of anticancer and other cytotoxic agents that may be used in combination with the inventive compounds include the following: epothilone derivatives as found in German Patent No. 4138042.8; WO 97/19086, WO 98/22461, WO 98/25929, WO 98/38192, WO 99/01124, WO 99/02224, WO 99/02514, WO 99/03848, WO 99/07692, WO 99/27890, WO 99/28324, WO 99/43653, WO 99/54330, WO 99/54318, WO 99/54319, WO 99/65913, WO 99/67252, WO 99/67253 and WO 00/00485; cyclin dependent kinase inhibitors as found in WO 99/24416; and prenyl-protein transferase inhibitors as found in WO 97/30992 and WO 98/54966.
  • The above other therapeutic agents, when employed in combination with the compounds of the present invention, may be used, for example, in those amounts indicated in the Physicians' Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art.
  • The inventive compounds have been tested in cell-cell assays and demonstrated activity consistent with inhibition of LFA-1 and/or ICAM-1.
  • Assays
  • H1-HeLa Adhesion Assay
  • H1-Hela cells were released from their growth flask using versene (Gibco, Grand Island, N.Y.). Following centrifugation, the cells were resuspended in growth medium: DMEM (Gibco), 10% fetal calf serum (Hyclone, Logan, Utah), 1% Pen-Strep (Gibco), and 1% L-glutamine (Gibco) and plated for growth at 5,000 cells/well in a 96-well plate.
  • The next day, HSB-2 cells were divided to 2×105/ml in growth medium: RPMI 1640 (Gibco), 10% FCS, 1% Pen-Strep, and 1% L-glutamine. The next day (day #3), the cells were centrifuged at 534×G for 8 minutes, washed, and resuspended in HBSS at 5×107/ml. Calcein-AM, 10 μM (Molecular Probes, Eugene, Oreg.) and 100 nM phorbol myristate acetate (SIGMA, St. Louis, Mo.) were added to the labeling and activation mix. Following incubation at 37° C. for 30 minutes, ten ml of HBSS was added and the cells centrifuged as above. The cell pellet was then resuspended and counted.
  • While the HSB-2 cells were labeling, the medium was aspirated from the H1-HeLa cells and the plates washed once with HBSS, followed by the addition of 50 μl of HBSS. An additional 50 μl of HBSS containing compound solution, DMSO, or anti-CD18 antibody was then added to each well. To the H1-HeLa cells were added 200,000 HSB-2 cells/well in 100 μl, followed by incubation in the dark for 30 minutes. The wells were then washed three times to remove the unbound cells. A fluorescence plate reader was then used to determine the number of bound HSB-2 cells. The percent inhibition due to the compound was calculated using the vehicle control as 0% inhibition and the antibody blocked adhesion as 100% inhibition.
  • HUVEC Adhesion Assay
  • On day 1, human umbilical vein endothelial cells (HUVEC) (passage 3, Clonetics, San Diego, Calif.) were placed into a T-75 flask containing EGM bulletkit media (Clonetics) for growth.
  • When the HUVEC were 90% confluent (typically day 4), 96-well tissue culture plates were coated with 100 μl/well of 2.5 μg/ml mouse Type IV collagen (Gibco) diluted in 0.1 M acetic acid. Following incubation for at least three hours, the collagen was removed and the plate washed three times with HBSS (Gibco). The HUVEC flask was trypsinized, and HUVEC were plated on the collagen coated wells at 1250 cells/200 μl/well for use four days later. Twenty hours prior to use, the medium was removed and cells were stimulated with 200 μl of 1 μg/ml lipopolysaccharide (LPS, Sigma, St. Louis, Mo.) in EGM. When the cells were 90% confluent (typically day 8), the LPS-containing medium was removed, the wells were washed with HBSS, and 50 μl of HBSS was added to the wells. An additional 50 μl containing compound solution, DMSO or blocking anti-CD18 was then added to each well.
  • On day 7, HSB-2 cells were then divided to 2×105/ml in RPMI 1640 (Gibco), 10% FCS (Hyclone, Logan, Utah), 1% Pen-Strep (Gibco), and 1% L-glutarmne (Gibco). The following day, the cells were centrifuged at 534×G for 8 minutes, washed, and resuspended in HBSS at 5×107/ml. For activation and labeling, calcein-AM, 10 μM (Molecular Probes, Eugene, Oreg.) and 100 nM phorbol myristate acetate (Sigma, St. Louis, Mo.) were added and the cells incubated at 37° C. for 30 minutes. Following the addition of ten ml of HBSS, the cells were centrifuged, resuspended, and counted.
  • To the HUVEC cells were added 200,000 labeled and activated HSB-2 cells/well in 100 μl, followed by incubation in the dark for 30 minutes. To remove unbound cells, the wells were washed three times with HBSS. A fluorescence plate reader was used to determine the number of HSB-2 cells bound. The percent inhibition due to the compound was calculated with the vehicle control set at 0% inhibition and the antibody-blocked adhesion set at 100% inhibition.
  • EXAMPLES
  • The following Examples illustrate embodiments of the inventive compounds and starting materials, and are not intended to limit the scope of the claims. For ease of reference, the following abbreviations are used herein:
  • Abbreviations
      • AlCl3=aluminum chloride
      • Ac2O=acetic anhydride
      • AcONa=sodium acetate
      • Bn=benzyl
      • Bu=butyl
      • t-Bu=tertiary butyl
      • Boc=tert-butyl oxycarbonyl
      • bp=boiling point
      • CDCl3=deutered chloroform
      • CH3CN=acetonitrile
      • DCM=dichloromethane
      • DMAP=4-dimethylaminopyridine
      • DCC=dicyclohexylcarbodiimide
      • DIPEA or DIEA=N,N-diisopropylethylamine
      • DME=1,2-dimethoxyethane
      • DMF=dimethyl formamide
      • EDCI=1-3-dimethylaminopropyl)-3-ethylcarbodiimide
      • Et2O=diethyl ether
      • HOBT=1-hydroxybenzotriazole
      • eq=equivalent(s)
      • h or hr=hour(s)
      • EtOAc=ethyl acetate
      • g=gram(s)
      • h or hr=hour(s)
      • HCl=hydrochloric acid
      • HPLC=high performance liquid chromatography
      • KOH=potassium hydroxide
      • K2CO3=potassium carbonate
      • l=liter
      • LiAlH4=lithium aluminum hydride
      • LDA=lithium diisopropyl amide
      • MeCN=acetonitrile
      • MeOH=methanol
      • mg(s)=milligram(s)
      • MgSO4=magnesium sulfate
      • NaH=sodium hydride
      • NaHCO3=sodium bicarbonate
      • Na2SO4=sodium sulfate
      • NaOH=sodium hydroxide
      • NMP=1-methyl-2-pyrrolidinone
      • pet. ether=petroleum ether
      • PBr3=phosphorus tribromide
      • (Ph3P)4Pd=tetrakis(triphenylphosphine)palladium(0)
      • PS=polystyrene
      • SiO2=silica
      • SOCl2=thionyl chloride
      • TEA (or Et3N)=triethylamine
      • THF=tetrahydrofuran
      • TFA=trifluoro acid acid
      • mg=milligram(s)
      • ml=milliliter
      • μl=microliter
      • mmol=millimole
      • μmol=micromole
      • mol=mole
      • mp=melting point
      • RT=room temperature
      • sat or sat'd=saturated
        Preparations
        Preparation 1
        (3R,6R,7aS)-6-tert-butoxy-3-(trichloromethyl)tetrahydro-1H-pyrrolo[1,2-c][1,3]oxazol-1-one
        Figure US20050119279A1-20050602-C00013
  • To a suspension of trans-3-t-butoxy-L-proline (1 g) (0.53 mmol) in 20 mL of acetonitrile was added anhydrous chloral 0.5 mL (1 eq.), and the mixture was allowed to stir at RT for 72 h. The remaining starting material was removed by filtration and the acetonitrile evaporated. The resulting white solid residue was stirred with isopropyl ether, filtered, and washed with pentane to afford the above titled compound (1.45 g). Mp=147° C. 1H NMR (CDCl3): 5.1 (1H,s), 4.20 (1H,dd), 4.10 (1H,dd), 3.13-3.32 (2H,m), 2.05-2.25 (2H,m), 1.20 (9H,s).
  • Preparation 2
  • (3R,6R,7aS)-6-tert-butoxy-7a-(4-Bromo-benzyl)-3-(trichloromethyl)tetrahydro-1H-pyrrolo[1,2-c][1,3]oxazol-1-one
    Figure US20050119279A1-20050602-C00014
  • To a suspension of (3R,6R,7aS)-6-tert-butoxy-3-(trichloromethyl) tetrahydro-1H-pyrrolo[1,2-c][1,3]oxazol-1-one (1.45 g) (Prep. 1) in 10 mL of dry THF at −78° C. was added 6.9 mL of a 1M solution of LDA in THF (1.5 eq). The reaction mixture was stirred for 1 h, and a solution of 4-bromobenzyl bromide (3.5 g) (3eq.) in 5 mL of THF was added. The reaction mixture was stirred for an additional hour at −70° C., allowed to warm to −20° C., then partitioned between EtOAc and water. The aqueous layer was extracted with another volume of EtOAc. The combined organic extracts were dried over MgSO4, filtered and evaporated under reduced pressure to afford a brown oil which was purified by SiO2 chromatography (eluent: pet. ether then pet. ether/DCM-50/50) to give the above-titled compound as a white solid (0.8 g). Mp=140° C. 1H NMR (CDCl3): 7.45 (2H,d), 7.25 (2H,d), 5.0 (1H,s), 3.95 (1H,m), 3.25 (1H,d), 3.10 (1H,dd), 2.95 (1H,d), 2.50 (1H,dd), 2.30 (1H,dd), 1.85 (1H,dd), 1.13 (9H,s).
  • Preparation 3
  • methyl (4R)-2-(4-bromobenzyl)-4-hydroxy-L-prolinate
    Figure US20050119279A1-20050602-C00015
  • To a suspension of (3R,6R,7aS)-6-tert-butoxy-7a-(4-Bromo-benzyl)-3-(trichloromethyl)tetrahydro-1H-pyrrolo[1,2-c][1,3]oxazol-1-one (0.4 g) (Prep. 2) in 10 mL of MeOH was added 1 mL of dry HCl in ethyl ether, and the resulting solution was refluxed for 13 hrs. The MeOH was evaporated, and the residue dissolved in DCM and washed with aqueous NaHCO3. The organic layer was dried over MgSO4, filtered, and evaporated under reduced pressure to afford the above-titled compound (0.170 g). 1H NMR (CDCl3): 7.40 (2H,d), 7.05 (2H,d), 4.25 (1H, m), 3.65 (3H,s), 3.15 (1H,d), 2.85-3.0 (2H,m), 2.45-2.6 (2H,m), 1.85 (1H,dd).
  • Preparation 4
  • (6R,7aS)-7a-(4-bromobenzyl)-2-(3,5-dichlorophenyl)-6-hydroxytetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione
    Figure US20050119279A1-20050602-C00016
  • To a solution of methyl (4R)-2-(4-bromobenzyl)-4-hydroxy-L-prolinate (0.170 g) (Prep. 3) in 10 mL of DMF was added K2CO3 (0.15 g). The reaction mixture was cooled to 0° C., treated with 3,5-dichlorophenyl isocyanate (0.116 g), then allowed to warm to RT and stirred overnight. The insoluble material was removed by filtration, the DMF evaporated, and the residue dissolved in DCM. The organic layer was washed with water, dried over MgSO4, and evaporated under reduced pressure. The resulting oily residue was purified over SiO2 isolute column (10 g) (eluent: DCM/acetone-95/5) to afford the above-titled compound (0.100 g) as a white solid. 1H NMR (CDCl3): 7.45 (2H,d), 7.30 (1H,m), 7.10 (2H,d), 6.85 (2H,d), 4.75 (1H,m), 4.10 (1H,dd), 3.45 (1H,d), 3.40 (1H,dd), 3.25 (1H,d), 2.35 (1H, dd), 2.20 (1H,dd).
  • Preparation 5
  • (4R)-1-{[(3,5-dichlorophenyl)amino]carbonyl}-4-hydroxy-L-proline
    Figure US20050119279A1-20050602-C00017
  • To a clear solution of L,trans-4-hydroxy-proline (10 g) (0.076 mol) and K2CO3 (15.7 g) (1.1 eq.) in 100 mL of water and 10 mL of THF, was added by portions 3,5-dichlorophenyl isocyanate (15.7 g) (1.1 eq.). The white suspension was stirred at RT overnight. The aqueous mixture was acidified with conc. HCl, ,then extracted with EtOAc. The organic extracts were combined, washed with water, dried over MgSO4, filtered, and the solvent removed in vacuo to afford the titled compound (19 g) (yield 78.5%).
  • 1H NMR (DMSOd6): 8.7 (1H,COOH), 7.65 (2H,d), 7.1 (1H,m), 5.2 (1H,NH), 4.4 (2H,m), 3.6 (1H,dd), 3.45 (1H,d), 2.1-2.25 (1H, m), 1.85-2.05 (1H,m).
  • Preparation 6
  • (6R,7aS)-2-(3,5-dichlorophenyl)-6-hydroxytetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione
    Figure US20050119279A1-20050602-C00018
  • (4R)-1-{[(3,5-dichlorophenyl)amino]carbonyl}-4-hydroxy-L-proline (6.8 g) (0.021 mol) (Preparation 5) was combined with 35 mL of 1N HCl and refluxed for 11 hrs. The organic oil was separated from the reaction mixture and dissolved in EtOAc. The organic layer was washed with water, dried over MgSO4, and evaporated under reduced pressure. The resulting residue was purified on a SiO2 column (eluent: DCM/acetone-90/10) to afford the titled compound as a white solid (3.2 g). 1H NMR (CDCl3): 7.4 (2H,d), 7.35 (1H,m), 4.8 (1H,m), 4.60 (1H,dd), 3.98 (1H,dd), 3.35 (1H,d), 2.39 (1H,dd), 2.05 (1H,d,OH), 1.80-1.95 (1H,m).
  • Preparation 7
  • (6R,7aS)-2-(3,5-dichlorophenyl)-1,3-dioxohexahydro- 1H-pyrrolo[1,2-c]imidazol-6-yl benzoate
    Figure US20050119279A1-20050602-C00019
  • To a solution of (6R,7aS)-2-(3,5-dichlorophenyl)-6-hydroxytetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione (1.2 g) (4mmol) (Preparation 6), dimethylaminopyridine (50 mg) (0.1 eq.) and benzoic acid (0.49 g) (1 eq.) in 10 mL of dry DCM at 0° C. was added DCC (0.9 g) (1 eq.) in a minimal volume of DCM. The reaction mixture was stirred to RT overnight. After filtering off the insoluble material, the reaction mixture is washed with a dilute NaHCO3 aqueous solution, with dilute HCl solution , with water, dried over MgSO4, and evaporated under reduced pressure to afford to afford the titled compound as a viscous oil (1.54 g). 1H NMR (CDCl3): 8.05 (2H,d), 7.65 (1H,m), 7.5 (1H,d), 7.43 (2H,d), 7.38 (1H,m), 5.81 (1H,t), 4.62 (1H,dd), 4.23 (1H,dd), 3.56 (1H,d), 2.68 (1H,dd), 2.05-2.20 (1H,m).
  • Preparation 8 and 9
  • (6R,7aS)-7a-(4-cyanobenzyl)-2-(3,5-dichlorophenyl)-1,3 -dioxohexahydro-1H-pyrrolo[1,2-c]imidazol-6-yl benzoate
    Figure US20050119279A1-20050602-C00020

    and
    (6R,7aR)-7a-(4-cyanobenzyl)-2-(3,5-dichlorophenyl)-1,3-dioxohexahydro-1H-pyrrolo[1,2-c]imidazol-6-yl benzoate
    Figure US20050119279A1-20050602-C00021
  • To a solution of (6R,7aS)-2-(3,5-dichlorophenyl)-1,3-dioxohexahydro-1H-pyrrolo[1,2-c]imidazol-6-yl benzoate (1.5 g) (3.7 mmol) (Preparation 7), and □-bromomethyl-benzonitrile (0.72 g) (1 eq.) in 15 mL of dry DMSO was added in one portion finely grinded KOH (0.25 g) (1 eq.). The reaction mixture was stirred for 45 min at RT, then diluted with 150 mL of tButyl methyl ether. The organic phase was washed with water, dried over MgSO4, filtered, and concentrated in vacuo. The residue afford the two diastereoisomers which were separated by column chromatography on silica (eluent: pet.ether-DCM 50/50).
  • Isomer 1 (6R,7aS)-7a-(4-cyanobenzyl)-2-(3,5-dichlorophenyl)-1,3-dioxohexahydro-1H-pyrrolo[1,2-c]imidazol-6-yl benzoate (0.52 g)
  • 1H NMR (CDCl3): 8.05 (2H, d), 7.45-7.68 (5H, m), 7.34 (2H, d), 7.27 (1H, m), 6.93 (2H, d), 5.78 (1H, t), 4.47 (1H,dd), 3.64 (1H, dd), 3.58 (2H,bs), 2.45-2.67 (2H, m). M+(509)
  • Isomer 2 (6R,7aR)-7a-(4-cyanobenzyl)-2-(3,5-dichlorophenyl)-1,3-dioxohexahydro-1H-pyrrolo[1,2-c]imidazol-6-yl benzoate (0.37 g)
  • 1H NMR (CDCl3): 7.82 (2H,d), 7.53-7.65 (3H,m), 7.35-7.45 (4H,m), 7.31 (1H,m), 6.75 (2H,d), 5.55 (1H,m), 4.36 (1H,dd), 3.32-3.42 (2H,d+dd), 3.06 (1H,d), 2.84 (1H,bd), 2.35 (1H,dd). M+(509)
  • Preparation 10
  • 4-[(6R,7aS)-2-(3,5-dichlorophenyl) tetrahydro-6-hydroxy-1,3-dioxo-1H-pyrrolo[1,2-c]imidazol-7a(5H)-yl]-benzonitrile
    Figure US20050119279A1-20050602-C00022
  • To a solution of (6R,7aS)-7a-(4-cyanobenzyl)-2-(3,5-dichlorophenyl)-1,3-dioxohexahydro-1H-pyrrolo[1,2-c]imidazol-6-yl benzoate (0.52 g) (1.02 mmol) (Preparation 8) in 1.5 mL of methanol and 4.5 mL of tBuOMe was added in one portion finely grinded KOH (0.057 g) (1 eq.). The reaction mixture was stirred for 5 min at RT, then quenched with 1 mL of 1N HCl, diluted with t-butyl methyl ether. The organic phase was washed with water, dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by column chromatography on silica (eluent: pet.ether-DCM 50/50) to afford the titled compound (0.2 g)
  • 1H NMR (CDCl3): 7.60 (2H,d), 7.30-7.35 (3H,m), 6.88 (2H,d), 4.82 (1H,m), 4.21 (1H,dd), 3.63 (1H,d), 3.45 (1H,d), 3.34 (1H,d), 2.29 (2H,m).
  • Preparation 11
  • (7aS)-7a-[(4-bromophenyl)methyl]-2-(3,5-dichlorophenyl)dihydro-1H-pyrrolo[1,2-c]imidazole-1,3,6(2H,5H)-trione
    Figure US20050119279A1-20050602-C00023
  • To a solution of (6R,7aS)-7a-(4-bromobenzyl)-2-(3,5-dichlorophenyl)-6-hydroxytetrahydro-1H-pyrrolo[1,2-c]imidazole-1,3(2H)-dione (Preparation 4), (0.2 g, 0.42 mmol) in dichloromethane (5 mL) was added Dess-Martin periodane reagent (0.216 g, 0.504 mmol) in one lot at room temperature. The reaction mixture was stirred at room temperature for one hour and partitioned between dichloromethane (15 mL) and 1N NaOH (15 mL). The dichloromethane layer was washed with brine (2×20 mL), dried over sodium sulfate and concentrated to yield the title compound as a foamy solid (0.185 g). Retention time=3.84 min; column: YMC S5 Combiscreen ODS 4.6×50 mm (4 min. gradient); Solvent A=10% MeOH, 90% H2O, 0.2% H3PO4; Solvent B=90% MeOH, 10% H2O and 0.2% H3PO4.
  • Preparation 12
  • 4-[[(7aS)-2-(3.5-dichlorophenyl)-tetrahydro-1,3,6-trioxo-1H-pyrrolo[1,2-c]imidazol-7a(5H)-yl]methyl-benzonitrile
    Figure US20050119279A1-20050602-C00024
  • This compound was obtained using the protocol outlined for preparation 11 starting from 4-[(6R,7aS)-2-(3,5-dichlorophenyl) tetrahydro-6-hydroxy-1,3-dioxo-1H-pyrrolo[1,2-c]imidazol-7a(5H)-yl]-benzonitrile (Preparation 10), (0.2 g, 0.42 mmol). Yield: 0.193 g. 1H NMR (CDCl3): 7.65 (2H, d), 7.25 -7.35 (3H, m), 6.88 (2H, d), 4.25 (1H, d), 3.6 (1H, d), 3.35 (1H, d), 3.1 (1H, d), 2.85 (2H, q).
  • EXAMPLES Example 1 and 2
  • (2R,5S,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione
    Figure US20050119279A1-20050602-C00025

    and
    (2R,5R,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione
    Figure US20050119279A1-20050602-C00026
  • To a solution of (7aS)-7a-[(4-bromophenyl)methyl]-2-(3,5-dichlorophenyl)dihydro-1H-pyrrolo[1,2-c]imidazole-1,3,6(2H,5H)-trione (preparation 11, 0.12 g, 0.26 mmol) and (S) lactamide (0.114 g, 1.3 mmol) in xylene (20 mL) was added para-toluenesulfonic acid (10 mgs) and the contents refluxed with the azeotropic removal of water for eighteen hours. The reaction mixture was cooled to room temperature, concentrated under reduced pressure and partitioned between dichloromethane (25 mL) and IN sodium hydroxide (10 mL). The dichloromethane layer was dried over sodium sulfate, concentrated and purified by silica gel chromatography using hexane/ethyl acetate as the eluent.
  • Isomer 1: (2R,5S,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione (35 mg).
  • 1H NMR (CDCl3): 8.0 (1H, brs), 7.4 (2H, d), 7.3 (1H, m), 7.0 (2H, d), 6.8 (2H, s), 4.45 (1H, m), 4.0 (1H, d), 3.6 (1H, d), 3.35 (1H, d), 3.1 (1H, d), 1.4 (3H, d).
  • Isomer 2: (2R,5R,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione (45 mg)
  • 1H NMR (CDCl3): 7.8 (1H, brs), 7.4 (2H, d), 7.3 (1H, m), 7.1 (2H, d), 6.8 (2H, s), 4.35 (1H, m), 4.0 (1H, d), 3.15 (1H, d), 3.0 (1H, d), 2.9 (1H, d), 2.65 (1H, d), 2.25 (1H, d), 1.4 (3H, d).
  • The following examples were synthesized employing the protocol outlined for the preparation of Examples 1 and 2.
    Starting Coupling Retention
    Ex Structure material partner timea
    3
    Figure US20050119279A1-20050602-C00027
    Figure US20050119279A1-20050602-C00028
    Figure US20050119279A1-20050602-C00029
    4
    Figure US20050119279A1-20050602-C00030
    Figure US20050119279A1-20050602-C00031
    Figure US20050119279A1-20050602-C00032
    5
    Figure US20050119279A1-20050602-C00033
    Figure US20050119279A1-20050602-C00034
    Figure US20050119279A1-20050602-C00035
    3.57 min
    6
    Figure US20050119279A1-20050602-C00036
    Figure US20050119279A1-20050602-C00037
    Figure US20050119279A1-20050602-C00038
    3.54
    7
    Figure US20050119279A1-20050602-C00039
    Figure US20050119279A1-20050602-C00040
    Figure US20050119279A1-20050602-C00041
    3.13
    8
    Figure US20050119279A1-20050602-C00042
    Figure US20050119279A1-20050602-C00043
    Figure US20050119279A1-20050602-C00044
    3.17

    aColumn conditions: YMC S5 Combiscreen ODS 4.6 × 50 nm (4 min. gradient); Solvent A = 10% MeOH, 90% H2O, 0.1% TFA; Solvent B = 90% MeOH, 10% H2O and 0.1% TFA.
  • Example 9
  • (2R,5R,7a′S)-2′-(3,5-dichlorophenyl)-7a′-[[4-(pyrimidin-4-yl)-phenyl)-methyl]-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione
    Figure US20050119279A1-20050602-C00045
  • To a mixture of (2S,5S,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione (example 3, 0.02 g, 0.037 mmol) and 3-pyridylboronic acid (0.010 g, 0.08 mmol) in dimethoxyethane (2 mL) was sequentially added potassium carbonate (0.009 g, 0.069 mmol), and tetrakis (triphenylphosphine)palladium(0) (0.004 g). The reaction mixture was purged with nitrogen and heated at 80° C. for eighteen hours, cooled to room temperature and filtered over a thin pad of celite. The celite pad was washed with dichloromethane (10 mL) and the filtrate was subjected to preparative HPLC (YMC 20×100 mm; Solvent A=10% MeOH, 90% H2O, 0.1% trifluoroacetic acid; Solvent B=90% MeOH, 10% H2O and 0.1% trifluoroacetic acid) to yield title compound as its trifluoroacetic acid salt. Yield: 2.8 mgs. Retention time=2.52 min. YMC ODS S5 C18 4.6×50 mm (4 min. gradient); Solvent A=10% MeOH, 90% H2O, 0.1% TFA; Solvent B=90% MeOH, 10% H2O and 0.1% TFA.
  • Example 10 and 11
  • ethyl (4′S,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate
    Figure US20050119279A1-20050602-C00046

    and
    ethyl (4′S,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate
    Figure US20050119279A1-20050602-C00047
  • To a solution of 4-[[(7aS)-2-(3.5-dichlorophenyl)-tetrahydro-1,3,6-trioxo-1H-pyrrolo[1,2-c]imidazol-7a(5H)-yl]methyl-benzonitrile (preparation 12, 0.32 g, 0.77 mmol) and (D) Cysteine ethyl ester, hydrochloride (0.14 g, 0.77 mmol) in toluene (5 mL) was added ET3N (1.21 ml, 1.54 mmol) then para-toluenesulfonic acid (10 mgs) and the contents refluxed with the azeotropic removal of water for five hours. The reaction mixture was cooled to room temperature, diluted with t-butyl methyl ether, washed with water, NaHCO3 solution, water then dried over sodium sulfate, concentrated and purified by silica gel chromatography using dichloromethane/acetone as the eluent.
  • Isomer 1: (ethyl (4′S,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate):
  • yield: 0.016 g. 1H NMR (CDCl3): 7.6 (2H, d), 7.35 (2H, d), 7.3 (1H, m), 6.75 (2H, m), 4.2 (2H, q), 3.95 (1H, d), 3.9-3.7 (1H, m), 3.45-3.25 (2H, m), 3.1-2.85 (3H, m), 2.7-2.3 (2H, m+NH), 1.3 (3H,t).
  • Isomer 2: (ethyl (4′S,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate):
  • yield: 0.016 g. 1H NMR (CDCl3): 7.55 (2H, d), 7.4-7.2 (3H, m), 6.8 (2H, m), 4.4-4.2 (3H, m), 4.0 (1H, m), 3.7-3.0 (5H, m), 2.8 (NH), 2.65-2.35 (2H, dd), 1.3 (3H, t)
  • Example 12 and 13
  • ethyl (4′R,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate
    Figure US20050119279A1-20050602-C00048

    and
    ethyl (4′R,6S,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo [1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate
    Figure US20050119279A1-20050602-C00049
  • To a solution of 4-[[(7aS)-2-(3.5-dichlorophenyl)-tetrahydro-1,3,6-trioxo-1H-pyrrolo[1,2-c]imidazol-7a(5H)-yl]methyl-benzonitrile (preparation 12, 0.10 g, 0.24 mmol) and (L) Cysteine ethyl ester, hydrochloride (0.045 g, 0.24 mmol) in toluene (5 mL) was added ET3N (0.06 ml, 0.48 mmol) then para-toluenesulfonic acid (10 mgs) and the contents refluxed with the azeotropic removal of water for five hours. The reaction mixture was cooled to room temperature, diluted with t-butyl methyl ether, washed with water, NaHCO3 solution, water then dried over sodium sulfate, concentrated to give an oil (0.40 g), (0.10 g) of which was purified by silica gel chromatography using dichloromethane/acetone as the eluent.
  • Isomer 1: (ethyl (4′R,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate):
  • yield: 0.066 g. 1H NMR (CDCl3): 7.65 (2H, d), 7.4 (2H, d), 7.3 (1H, m), 6.8 (2H, m), 4.25 (2H, q), 4.1 (1H, d), 3.9-3.75 (1H, m), 3.5-3.35 (2H, m), 3.3 (1H, d), 3.1-2.95 (2H, m), 2.75 (1H, d), 2.5 (NH, d), 2.15 (1H, d), 1.3 (3H, t).
  • Isomer 2: (ethyl (4′R,6S,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate):
  • yield: 0.025 g. 1H NMR (CDCl3): 7.6 (2H, d), 7.35 (2H, d), 7.3 (1H, m), 6.85 (2H, m), 4.25 (2H,q), 4.1 (1H, d), 4.0-3.85 (1H, m), 3.55-3.45 (2H, m), 3.3 (2H, s), 3.159 (1H, dd), 3.0 (NH, d), 2.95 (1H, d), 2.55 (1H, d), 1.3 (3H, t).
  • Example 14
  • (±) ethyl (4′S,7aS)-3′-acetyl-7a-ethyl-2-methyl-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate
    Figure US20050119279A1-20050602-C00050
  • To a solution of ethyl (4′R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate (examples 12-13, crude mixture of isomers, 0.30 g, 0.58 mmol) and ET3N (0.10 ml, 0.6 mmol)) in acetonitrile (3 mL) was added acetyl chloride (0.05 ml) and the contents stirred one night at RT. The reaction mixture was concentrated under reduced pressure and partitioned between dichloromethane (25 mL) and 1N Hydrochloric acid (10 mL). The dichloromethane layer was washed with 1N Sodium Bicarbonate, water then dried over sodium sulfate, concentrated and purified by silica gel chromatography using cyclohexane/ethyl acetate as the eluent, diluted with t-butyl methyl ether, washed with water, NaHCO3 solution, water then dried over sodium sulfate, concentrated and purified by silica gel chromatography using dichloromethane/acetone as the eluent. yield: 0.016 g. 1H NMR (CDCl3): 7.6 (2H, d), 7.35 (2H, d), 7.3 (1H, m), 6.8 (2H, m), 4.9 (1H, m), 4.75 (1H, d), 4.3 (2H, q), 4.05 (1H, d), 3.5-3.2 (5H, m), 2.65 (1H, d), 2.15 (3H, s), 1.3 (3H, t).
  • Example 15
  • (±) ethyl (4′S,7aS)-7a-(4-bromophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate
    Figure US20050119279A1-20050602-C00051
  • To a solution (7aS)-7a-[(4-bromophenyl)methyl]-2-(3,5-dichlorophenyl)dihydro-1H-pyrrolo[1,2-c]imidazole-1,3,6(2H,5H)-trione (preparation 11, 1.0 g, 2.0 mmol) and (D) Cysteine ethyl ester, hydrochloride (0.40 g, 2.0 mmol) in toluene (10 mL) was added Et3N (0.60 ml, 4.0 mmol) then para-toluenesulfonic acid (10 mg) and the contents refluxed with the azeotropic removal of water for seven hours. The reaction mixture was cooled to room temperature, diluted with t-butyl methyl ether, washed with water, NaHCO3 solution, water then dried over sodium sulfate, concentrated and purified by silica gel chromatography using Ethyl acetate/Cyclohexane as the eluent. yield: 0.40 g. 1H NMR (CDCl3): 7.4 (2H, d), 7.25 (1H, m), 7.1 (2H, d), 6.75 (2H, m), 4.15 (2H, q), 3.9 (1H, d), 3.82-3.76 (1H, m), 3.35 (1H,

Claims (21)

1. A compound of formula (I),
Figure US20050119279A1-20050602-C00052
its stereoisomers, or a pharmaceutically-acceptable salt, hydrate, or prodrug thereof, in which:
K and L are independently O or S;
Q is a bond, —C(═O)— or branched or straight chain C1-6alkylene optionally substituted with one to two R4;
Ar is aryl or heteroaryl;
Y is a bond or —C(R6aR7a)—;
J1 is —N(R5)— or —C(R6bR7b)—;
J2 is —N(R5)— or —C(R6cR7c)—;
J3 is N(R5) or C(R7dR7d);
provided that only one of J1, J2 and J3 may be —N(R5)—, so that ring A is a five-to six-membered cycloalkyl or heterocyclo ring having from 0 to 2 heteroatoms; R1 is N or C(R9);
R2 and R3 are independently selected from hydrogen, halogen, nitro, cyano, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —SR12, —OR12, —NR12R13, —CO2R12, —C(═O)R12, —C(═O)NR12R13, aryl, heterocyclo, cycloalkyl, and heteroaryl;
R4 is selected from OH, O(C1-6alkyl), halogen, cyano, CF3, OCF3, NH2, NH(C1-6alkyl), and N(C1-6alkyl)2;
R5 is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —NR14R15, —C(═O)R14, —CO2R14, —C(═O)NR14R15, —S(O)pR15a, —SO2NR14R15, aryl, heterocyclo, cycloalkyl, and heteroaryl; or when R5 is joined to atom J1, J2 or J3, R5 may be taken together with one of R6a, R6b R6c or R6d attached to an adjacent atom of ring A to form a fused heterocyclo or heteroaryl ring;
at least one of R6a with R7a, or R6b with R7b, or R6c with R7c are taken together to form a spiro-cycloalkyl or spiro-heterocyclo ring; provided that the spiro-heterocyclo ring is not C2-3 alkylenedioxy;
R6a, R6b, R6c, R6d, R7a, R7b, R7c, and R7d which do not form a spiro-cycloalkyl or spiro-heterocyclo ring are independently selected from hydrogen, halogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, nitro, cyano, —SR16, —OR16, —NR16R17, —C(═O)R16, —CO2R16, —C(═O)NR16R17, —NR16C(═O)R17, —NR16C(═O)OR17, —S(O)qR17a, —NR16SO2R17a, —SO2NR16R17, aryl, heterocyclo, cycloalkyl, and heteroaryl;
or R6a, R6b, R6c, R6d, R7a, R7b, R7c, and R7d which do not form a spiro-cycloalkyl or spiro-heterocyclo ring and which are attached to the same carbon atom may be taken together to form a keto group;
or R6a, R6b, R6c, R6d, R7a, R7b, R7c, and R7d which do not form a spiro-cycloalkyl or spiro-heterocyclo ring and which are attached to adjacent carbon atoms may be taken together to form a fused benzo, cycloalkyl, heterocyclo, or heteroaryl ring;
R9 is selected from hydrogen, halogen, nitro, cyano, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —SR18, —OR18, —NR18R19, —CO2R18, —C(═O)R18, —C(═O)NR18R19, aryl, heterocyclo, cycloalkyl, and heteroaryl;
R12, R13, R14, R15, R16, R17, R18, and R19 (i) are selected independently of each from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo; or (ii) any two of R12, R13, R14, R15, R16, R17, R18, and R19 when attached to the same nitrogen atom may be taken together to form a heteroaryl or heterocyclo ring, with the remainder of R10, R11, R12, R13, R14, R15, R16, R17, R18, and R19 being selected independently from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo;
R15a, and R17a are independently selected from alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo;
p is 1, 2, or 3; and
q is 1, 2, or 3.
2. A compound according to claim 1, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, wherein:
K iand L are both O;
Q is a straight chain C1-6alkylene;
Ar is phenyl optionally substituted one to three R20;
R2 and R3 are selected from halogen, (C1-6)alkyl, cyano, halo(C1-6)alkyl, halo(C1-6)alkoxy, nitro, phenyloxy, benzyloxy, and phenylthio;
R20 at each occurrence is independently selected from halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, —C(═O) (C1-6)alkyl, —C(═O)NH(CH2)rCO2H, —C(═O)NH(CH2)rCO2(C1-6alkyl), and S(O)2(C1-6alkyl); or from phenyl, benzyl, phenyloxy, benzyloxy and heteroaryl in turn optionally substituted with one to two of halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, and/or —C(═O) (C1-6)alkyl; or alternatively, two R20 groups join together with each other to form a fused benzo ring; and
r is 1, 2, 3, or 4.
3. A compound according to claim 2, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, wherein Q-Ar together form:
Figure US20050119279A1-20050602-C00053
wherein
R20a and R20b are independently selected from halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, —C(═O) (C1-6)alkyl, —C(═O)NH(CH2)rCO2H, —C(═O)NH(CH2)rCO2(C1-6alkyl), and S(O)2(C1-6alkyl); or from phenyl, benzyl, phenyloxy, benzyloxy and heteroaryl in turn optionally substituted with one to two of halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —NH2, —NH(C1-6alkyl), —N(C1-6alkyl)2, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, and/or —C(═O) (C1-6)alkyl; or alternatively, two R20b groups join together with each other or one R20b joins together with R20a to form a fused benzo ring;
n is 0, 1, or 2; and
r is 1, 2, 3, or 4.
4. A compound according to claim 1, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, wherein
Y is a bond;
J3 is —CHR6b—;
J2 is is —C(R6cR7c)-wherein R6c with R7, are taken together to form a spiro-cycloalkyl or spiro-heterocyclo ring;
J3 is CH(R6d);
R6b, and R6d are independently selected from
a) hydrogen, halogen, and cyano;
b) —SR16, —OR16, —NR16R17, —C(═O)R16, —CO2R16, —C(═O)NR16R17, —NR16C(═O)R17, —NR16C(═O)OR17, —S(O)qR17a, —NR16SO2R17a, and —SO2NR16R17; and
c) C1-6alkyl, phenyl, four to seven membered heterocyclo, C3-7cycloalkyl, and five to six membered heteroaryl, each of which in turn is optionally substituted with one to two groups selected from R22;
R16 and R17 are selected independently from hydrogen, C1-6alkyl, phenyl, four to seven membered heterocyclo, C3-7cycloalkyl, and five to six membered heteroaryl, each of which in turn is optionally substituted with one to two groups selected from R23;
R17a is C1-6alkyl, phenyl, four to seven membered heterocyclo, C3-7cycloalkyl, five to six membered heteroaryl each of which is optionally substituted with one to two groups selected from R23; and
R22 and R23 are at each occurrence selected independently from halogen, cyano, C1-6alkyl, hydroxy, trifluoromethyl, trifluoromethoxy, —O(C1-6alkyl), —C(═O)H, —C(═O)(C1-6alkyl), —CO2H, —CO2(C1-6alkyl), —C(═O)NH2, —C(═O)NH2, —C(═O)NH(C1-6alkyl), —C(═O)N(C1-6alkyl)2, —NH2, —NH(C1-6alkyl), —N(C1-6alkyl)2, hydroxy(C1-6)alkyl, methoxy(C1-6)alkyl, ethoxy(C1-6)alkyl, amino(C1-6)alkyl, and halo(C1-6)alkyl.
5. The compound according to claim 1, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, in which
R1 is C(R9); and
R2 and R3 are independently halogen.
6. A compound having the formula (Ia)
Figure US20050119279A1-20050602-C00054
or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, in which
K and L are independently O or S;
Q is a bond, —C(═O)— or branched or straight chain C1-6alkylene optionally substituted with one to two R4;
Ar is aryl or heteroaryl;
Y is a bond or —C(R6aR7a)—;
J1 is —N(R5)— or —C(R6bR7b)—;
J3 is —N(R5)— or —C(R6dR7d)—;provided, that only one of J1 and J3 may be —N(R5)—, so that ring A is a five-to-six membered cycloalkyl or heterocyclo ring having from 0 to 2 heteroatoms;
Z1 and Z2 are independently NR25, S or O; so that Z forms a spiro heterocyclic ring provided that Z is not a C2-3 alkylenedioxy ring and that only one of Z1 and Z2 may be NR25;
R1 is N or C(R9);
R2 and R3 are independently selected from hydrogen, halogen, nitro, cyano, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —SR12, —OR12, —NR12R13, —CO2R12, —C(═O)R12, —C(═O)NR12R13, aryl, heterocyclo, cycloalkyl, and heteroaryl;
R4 is selected from OH, O(C1-6alkyl), halogen, cyano, CF3, OCF3, NH2, NH(C1-6alkyl), and N(C1-6alkyl)2;
R5 is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —NR14R15, —C(═O)R14, —CO2R14, —C(═O)NR14R15, —S(O)pR15a, —SO2NR14R15, aryl, heterocyclo, cycloalkyl, and heteroaryl; or when R5 is joined to atom J1, J3 or Y, R5 may be taken together with one of R6a, R6b or R6d attached to an adjacent atom of ring A to form a fused heterocyclo or heteroaryl ring;
R6a, R6b, R6d, R7a, R7b, R7b and R7d are independently selected from hydrogen, halogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, nitro, cyano, —SR16, —OR16, —NR16R17, —C(═O)R16, —CO2R16, —C(═O)NR16R17, —NR16C(═O)R17, —NR16C(═O)OR17, —S(O)qR17a, —NR16SO2R17a, —SO2NR16R17, aryl, heterocyclo, cycloalkyl, and heteroaryl;
R9 is selected from hydrogen, halogen, nitro, cyano, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —SR18, —OR18, —NR18R19, —CO2R18, —C(═O)R18, —C(═O)NR18R19, aryl, heterocyclo, cycloalkyl, and heteroaryl;
R12, R13, R14, R15, R16, R17, R18, and R19 (i) are selected independently of each other from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo; or (ii) any two of R12, R13, R14, R15, R16, R17, R18, and R19 when attached to the same nitrogen atom (as in NR12R13, NR14R15, NR16R17, or NR18R19) may be taken together to form a heteroaryl or heterocyclo ring, with the remainder of R12, R13, R14, R15, R16, R17, R18, and R19 being selected independently from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo;
R11a, R15a, and R17a are independently selected from alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo;
R25 is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —NR26R27, —C(═O)R26, —CO2R26, —C(═O)NR26R27, —S(O)pR27, —SO2NR26R27, aryl, heterocyclo, cycloalkyl, and heteroaryl;
R26, and R27 are selected independently of each other from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo;
p is 0, 1, 2, or 3; and
q is 1, 2, or 3.
7. A compound according to claim 6, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, in which
Y is a bond; and
Q-Ar together form:
Figure US20050119279A1-20050602-C00055
wherein
R20a and R20b are independently selected from halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, —C(═O) (C1-6)alkyl, —C(═O)NH(CH2)rCO2H, —C(═O)NH(CH2)rCO2(C1-6alkyl), and S(O)2(C1-6alkyl); or from phenyl, benzyl, phenyloxy, benzyloxy and heteroaryl in turn optionally substituted with one to two of halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —NH2, —NH(C1-6alkyl), —N(C1-6alkyl)2, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, and/or —C(═O) (C1-6)alkyl; or alternatively, two R20b groups join together with each other or one R20b joins together with R20a to form a fused benzo ring;
n is 0, 1, or 2; and
r is 1, 2, 3, or 4.
8. A compound according to claim 6, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, in which:
R2 and R3 are both halogen; and
R1 is CH.
9. A compound according to claim 6, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, in which
Y is a bond;
J1 is —C(R6bR7b)—; and
J3 is —C(R6dR7d)—.
10. A compound according to claim 6, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, in which one of Z1 or Z2 is NR25.
11. A compound of formula (Ib),
Figure US20050119279A1-20050602-C00056
(Ib)
its stereoisomers, or a pharmaceutically-acceptable salt, hydrate, solvate, or prodrug thereof, in which:
one of Z1 or Z2 is NR25 and the other of Z1 or Z2 is O or S;
R1 is N or C(R9);
R2 and R3 are independently selected from halogen, (C1-6)alkyl, cyano, halo(C1-6)alkyl, halo(C1-6)alkoxy, nitro, phenyloxy, benzyloxy, and phenylthio;
R6 is selected from (a) halogen, nitro, and cyano; or from (b) —SR16, —OR16, —NR16R17, —C(═O)R16, —CO2R16, —C(═O)NR16R17, —NR16C(═O)R17, —NR16C(═O)OR17, —S(O)qR17a, —NR16SO2R17a, and —SO2NR16R17; or from (c) alkyl, alkenyl, aryl, heterocyclo, cycloalkyl, and heteroaryl, in turn optionally substituted with one to two groups selected from R22; and/or (d) two R6 groups taken together form keto (═O), with the remainder of the R6 groups selected from (a), (b), and (c);
R16 and R17 are selected independently of each other from hydrogen, C1-6alkyl, phenyl, four to seven membered heterocyclo, C3-7cycloalkyl, and five to six membered heteroaryl, each of which in turn is optionally substituted with one to two groups selected from R23;
R17a is C1-6alkyl, phenyl, four to seven membered heterocyclo, C3-7cycloalkyl, five to six membered heteroaryl each of which is optionally substituted with one to two groups selected from R23;
R20a and R20b are independently selected from halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, —C(═O) (C1-6)alkyl, —C(═O)NH(CH2)rCO2H, —C(═O)NH(CH2)rCO2(C1-6alkyl), and —S(O)2(C1-6alkyl); or from phenyl, benzyl, phenyloxy, benzyloxy and heteroaryl in turn optionally substituted with one to two of halogen, C1-6alkyl, hydroxy, (C1-6)alkoxy, halo(C1-6)alkyl, halo(C1-6)alkoxy, cyano, nitro, —NH2, —NH(C1-6alkyl), —N(C1-6alkyl)2, —CO2H, —C(═O)H, —CO2(C1-6)alkyl, and/or —C(═O) (C1-6)alkyl; or alternatively, two R20b groups join together with each other or one R20b joins together with R20a to form a fused benzo ring;
R22, R23 and R24 are at each occurrence selected independently from halogen, cyano, nitro, C1-6alkyl, hydroxy, trifluoromethyl, trifluoromethoxy, —O(C1-6alkyl), —C(═O)H, —C(═O)(C1-6alkyl), —CO2H, —CO2(C1-6alkyl), —C(═O)NH2, —C(═O)NH2,—C(═O)NH(C1-6alkyl), —C(═O)N(C1-6alkyl)2, —NH2, —NH(C1-6alkyl), —N(C1-6alkyl)2, hydroxy(C1-6)alkyl, methoxy(C1-6)alkyl, ethoxy(C1-6)alkyl, amino(C1-6)alkyl, and halo(C1-6)alkyl;
R25 is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, —NR26R27, —C(═O)R26, —CO2R26, —C(═O)NR26R27, —S(O)pR27, —SO2NR26R27, aryl, heterocyclo, cycloalkyl, and heteroaryl;
R26 and R27 are selected independently of each other from hydrogen, alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl, and heterocyclo
n, s and and p are independently 0, 1, or 2;
q is 1, 2, or 3; and
r is 0, 1, 2, 3 or 4.
12. A compound according to claim 11, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, in which R2 and R3 are independently halogen.
13. A compound according to claim 11, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, in which R20a is cyano, halogen, aryl, or heteroaryl.
14. A compound according to claim 11, or a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer thereof, in which
R1 is CH;
R2 and R3 are independently halogen;
R6 is hydrogen, C1-6alkyl, substituted C1-6alkyl, —C(═O)R26 or CO2R26;
R25 is hydrogen, C1-6alkyl, substituted C1-6alkyl, or —C(═O)R26; and
n is 0.
15. A compound having the formula,
(i) (2R,5S,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
(2R,5R,7a′S)-7a′-[(4-bromophenyl)-methyl]-2′-(3,5-dichlorophenyl)-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
(2R,5R,7a′S)-7′a-[(4-Bromophenyl)methyl]-2′-(3,5-dichlorophenyl)dihydro-5-methylspiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
(2S,5R,7a′S)-7′a-[(4-Bromophenyl)methyl]-2′-(3,5-dichlorophenyl)dihydro-5-methylspiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo [1,2-c]imidazole]-1′,3′,4(2′H)-trione;
(2R,7a′S)-7′a-[(4-Bromophenyl)methyl]-2′-(3,5-dichlorophenyl)dihydrospiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
(2S ,7a′S)-7′a-[(4-Bromophenyl)methyl]-2′-(3,5-dichlorophenyl)dihydrospiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
4-[[(2R,5R,7a′S)-2′-(3,5-Dichlorophenyl)dihydro-5-methyl-1′,3′,4-trioxospiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo[1,2-c]imidazol]-7′a(7′H)-yl]methyl]benzonitrile;
4-[[(2S,5R,7a′S)-2′-(3,5-Dichlorophenyl)dihydro-5-methyl-1′,3′,4-trioxospiro[oxazolidine-2,6′(5′H)-[1H]pyrrolo[1,2-c]imidazol]-7′a(7′H)-yl]methyl]-benzonitrile;
(2R,5R,7a′S)-2′-(3,5-dichlorophenyl)-7a′-[[4-(pyrimidin-4-yl)-phenyl)-methyl]-5-methyldihydro-1′H,4H-spiro[1,3-oxazolidine-2,6′-pyrrolo[1,2-c]imidazole]-1′,3′,4(2′H)-trione;
ethyl (4′S,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate;
ethyl (4′S,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate;
ethyl (4′R,6R,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate;
ethyl (4′R,6S,7aS)-7a-(4-cyanophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]4′-carboxylate;
(±) ethyl (4′S,7aS)-3′-acetyl-7a-ethyl-2-methyl-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate; or
(±) ethyl (4′S,7aS)-7a-(4-bromophenyl)-2-(3,5-dichlorophenyl)-1,3-dioxotetrahydro-1H-spiro[pyrrolo[1,2-c]imidazole-6,2′-[1,3]thiazolidine]-4′-carboxylate; or
(ii) a pharmaceutically-acceptable salt, hydrate, prodrug, or stereoisomer of (i).
16. A pharmaceutical composition comprising at least one compound according to claim 1 and a pharmaceutically acceptable carrier or diluent.
17. A pharmaceutical composition comprising at least one compound according to claim 16 and a pharmaceutically acceptable carrier or diluent.
18. A method of inhibiting an LFA-1/ICAM-associated condition in a mammal comprising administering to the mammal a therapeutically-effective amount of a compound according to claim 1.
19. The method of claim 18 wherein the LFA-1/ICAM-associated condition is an inflammatory or immune disease.
20. The method of claim 19 in which LFA-1/ICAM-associated condition is selected from acute or chronic graft vs host reactions, acute or chronic transplant rejection, multiple sclerosis, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, osteoporosis, diabetes, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, Crohn's disease, ulcerative colitis, Alzheimer's disease, shock, ankylosing spondylitis, gastritis, conjunctivitis, pancreatis, multiple organ injury syndrome, myocardial infarction, atherosclerosis, stroke, reperfusion injury, acute glomerulonephritis, vasculitis, thermal injury, necrotizing enterocolitis, granulocyte transfusion associated syndrome, Sjogren's syndrome, eczema, atopic dermatitis, contact dermatitis, urticaria, schleroderma, psoriasis, asthma, pulmonary fibrosis, allergic rhinitis, oxygen toxicity, emphysema, chronic bronchitis, acute respiratory distress syndrome, chronic obstructive pulmonary disease (COPD), hepatitis B, hepatitis C, organ-tissue autoimmune disease, autoimmune thyroiditis, uveitis, systemic lupus erythematosis, Addison's disease, autoimmune polyglandular disease, and Grave's disease.
21. The method of claim 20 in which the inflammatory or immune disease is selected from acute or chronic transplant rejection, rheumatoid arthritis, osteoarthritis, diabetes, asthma, inflammatory bowel disease, psoriasis, and chronic obstructive pulmonary disease.
US10/957,255 2003-10-02 2004-10-01 Spiro-cyclic compounds useful as anti-inflammatory agents Active 2025-05-31 US7199125B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/957,255 US7199125B2 (en) 2003-10-02 2004-10-01 Spiro-cyclic compounds useful as anti-inflammatory agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US50816503P 2003-10-02 2003-10-02
US10/957,255 US7199125B2 (en) 2003-10-02 2004-10-01 Spiro-cyclic compounds useful as anti-inflammatory agents

Publications (2)

Publication Number Publication Date
US20050119279A1 true US20050119279A1 (en) 2005-06-02
US7199125B2 US7199125B2 (en) 2007-04-03

Family

ID=34622918

Family Applications (1)

Application Number Title Priority Date Filing Date
US10/957,255 Active 2025-05-31 US7199125B2 (en) 2003-10-02 2004-10-01 Spiro-cyclic compounds useful as anti-inflammatory agents

Country Status (1)

Country Link
US (1) US7199125B2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007047146A2 (en) * 2005-10-11 2007-04-26 Intermune, Inc. Inhibitors of viral replication
GB2450771A (en) * 2007-04-12 2009-01-07 Angeletti P Ist Richerche Bio Heterocyclic compounds and their use as inhibitors of HCV polymersases for the treatment of HCV
US20090047246A1 (en) * 2007-02-12 2009-02-19 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
WO2009054914A1 (en) 2007-10-19 2009-04-30 Sarcode Corporation Compositions and methods for treatment of diabetic retinopathy
US20090170770A1 (en) * 2007-11-06 2009-07-02 Ali Hafezi-Moghadam Methods and compositions for treating conditions associated with angiogenesis using a vascular adhesion protein-1 (vap 1) inhibitor
US9216174B2 (en) 2003-11-05 2015-12-22 Sarcode Bioscience Inc. Modulators of cellular adhesion
JP2017524732A (en) * 2014-06-20 2017-08-31 ラトガース,ザ ステート ユニバーシティ オブ ニュー ジャージー How to treat lung inflammation
US10603354B2 (en) 2009-10-13 2020-03-31 Rutgers, The State University Of New Jersey Treatment and diagnosis of inflammatory disorders and HIV

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010141330A1 (en) 2009-06-02 2010-12-09 Boehringer Ingelheim International Gmbh DERIVATIVES OF 6,7-DIHYDRO-5H-IMIDAZO[1,2-a]IMIDAZOLE-3-CARBOXYLIC ACID AMIDES

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3941744A (en) * 1971-06-05 1976-03-02 Sankyo Company Limited Piperidine derivatives and their use as stabilizers
US4066615A (en) * 1971-06-05 1978-01-03 Sankyo Company Limited Polymer compositions containing piperidine derivatives as stabilizers
US4241208A (en) * 1971-06-05 1980-12-23 Sankyo Company Limited Piperidine derivatives
US6022875A (en) * 1997-07-31 2000-02-08 Gruenenthal Gmbh Use of substituted 2,4-imidazolidinedione compounds as analgesics
US20020143035A1 (en) * 2000-12-01 2002-10-03 Michele Launay Hydantoin compounds useful as anti-inflammatory agents
US20030232817A1 (en) * 2002-05-29 2003-12-18 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful for the treatment of inflammatory disease
US6897225B1 (en) * 1999-10-20 2005-05-24 Tanabe Seiyaku Co., Ltd. Inhibitors of αLβ2 mediated cell adhesion
US20060074099A1 (en) * 2004-10-01 2006-04-06 Delmonte Albert J Crystalline forms and process for preparing spiro-hydantoin compounds
US20060148836A1 (en) * 2004-12-14 2006-07-06 Dhar T G M Pyridyl-substituted spiro-hydantoin compounds and use thereof

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2278547A1 (en) 1997-03-03 1998-09-11 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
WO1999020618A1 (en) 1997-10-21 1999-04-29 Active Biotech Ab Thiadiazoles amides useful as antiinflammatory agents
AU9802198A (en) 1997-10-21 1999-05-10 Pharmacia & Upjohn Company Antiinflammatory thiadiazolyl ureas which act as lfa-1 and mac-1 inhibitors
ATE353640T1 (en) 1998-03-27 2007-03-15 Genentech Inc ANTAGONISTS FOR THE TREATMENT OF CD11/CD18 ADHESION RECEPTOR-MEDIATED DISEASES
WO2001007044A1 (en) 1999-07-21 2001-02-01 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
US6350763B1 (en) 1999-07-21 2002-02-26 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammation disease
US6492408B1 (en) 1999-07-21 2002-12-10 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
ATE396722T1 (en) 1999-07-21 2008-06-15 Boehringer Ingelheim Pharma SMALL MOLECULES FOR THE TREATMENT OF INFLAMMATORY DISEASES
US6365615B1 (en) 1999-07-21 2002-04-02 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
JP3943394B2 (en) 1999-10-20 2007-07-11 田辺製薬株式会社 αLβ2-mediated cell adhesion inhibitor
WO2001045704A1 (en) 1999-12-21 2001-06-28 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
CA2413683A1 (en) 2000-06-28 2002-01-03 Bristol-Myers Squibb Company Fused cyclic compounds as modulators of nuclear hormone receptor function
US20030114420A1 (en) 2000-06-28 2003-06-19 Salvati Mark E. Fused cyclic modulators of nuclear hormone receptor function
JP4109890B2 (en) 2001-04-19 2008-07-02 田辺三菱製薬株式会社 Pharmaceutical composition
NZ531870A (en) 2001-10-01 2005-08-26 Bristol Myers Squibb Co Spiro-hydantoin compounds useful as anti-inflammatory agents
TW200303200A (en) 2002-02-07 2003-09-01 Tanabe Seiyaku Co Inhibitors of α L β 2 integrin mediated cell adhesion

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3941744A (en) * 1971-06-05 1976-03-02 Sankyo Company Limited Piperidine derivatives and their use as stabilizers
US4066615A (en) * 1971-06-05 1978-01-03 Sankyo Company Limited Polymer compositions containing piperidine derivatives as stabilizers
US4241208A (en) * 1971-06-05 1980-12-23 Sankyo Company Limited Piperidine derivatives
US6022875A (en) * 1997-07-31 2000-02-08 Gruenenthal Gmbh Use of substituted 2,4-imidazolidinedione compounds as analgesics
US6897225B1 (en) * 1999-10-20 2005-05-24 Tanabe Seiyaku Co., Ltd. Inhibitors of αLβ2 mediated cell adhesion
US20020143035A1 (en) * 2000-12-01 2002-10-03 Michele Launay Hydantoin compounds useful as anti-inflammatory agents
US6710064B2 (en) * 2000-12-01 2004-03-23 Bristol-Myers Squibb Co. Hydantoin compounds useful as anti-inflammatory agents
US20030232817A1 (en) * 2002-05-29 2003-12-18 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful for the treatment of inflammatory disease
US20060074099A1 (en) * 2004-10-01 2006-04-06 Delmonte Albert J Crystalline forms and process for preparing spiro-hydantoin compounds
US20060148836A1 (en) * 2004-12-14 2006-07-06 Dhar T G M Pyridyl-substituted spiro-hydantoin compounds and use thereof

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9248126B2 (en) 2003-11-05 2016-02-02 Sarcode Bioscience Inc. Modulators of cellular adhesion
US9216174B2 (en) 2003-11-05 2015-12-22 Sarcode Bioscience Inc. Modulators of cellular adhesion
WO2007047146A2 (en) * 2005-10-11 2007-04-26 Intermune, Inc. Inhibitors of viral replication
WO2007047146A3 (en) * 2005-10-11 2007-11-01 Intermune Inc Inhibitors of viral replication
JP2009513576A (en) * 2005-10-11 2009-04-02 インターミューン・インコーポレーテッド Viral replication inhibitors
US20090099186A1 (en) * 2005-10-11 2009-04-16 Leonid Beigelman Inhibitors of viral replication
US20090047246A1 (en) * 2007-02-12 2009-02-19 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
GB2450771A (en) * 2007-04-12 2009-01-07 Angeletti P Ist Richerche Bio Heterocyclic compounds and their use as inhibitors of HCV polymersases for the treatment of HCV
US20090155176A1 (en) * 2007-10-19 2009-06-18 Sarcode Corporation Compositions and methods for treatment of diabetic retinopathy
WO2009054914A1 (en) 2007-10-19 2009-04-30 Sarcode Corporation Compositions and methods for treatment of diabetic retinopathy
US10960087B2 (en) 2007-10-19 2021-03-30 Novartis Ag Compositions and methods for treatment of diabetic retinopathy
EP3167886A1 (en) 2007-10-19 2017-05-17 SARcode Bioscience Inc. Compositions and methods for treatment of macular edema
EP3797775A1 (en) 2007-10-19 2021-03-31 Novartis AG Compositions and methods for treatment of diabetic retinopathy
US20090170770A1 (en) * 2007-11-06 2009-07-02 Ali Hafezi-Moghadam Methods and compositions for treating conditions associated with angiogenesis using a vascular adhesion protein-1 (vap 1) inhibitor
US10603354B2 (en) 2009-10-13 2020-03-31 Rutgers, The State University Of New Jersey Treatment and diagnosis of inflammatory disorders and HIV
US10993989B2 (en) 2009-10-13 2021-05-04 Rutgers, The State University Of New Jersey Treatment and diagnosis of inflammatory disorders and HIV
US11612636B2 (en) 2009-10-13 2023-03-28 Rutgers, The State University Of New Jersey Method of treating autoimmune inflammatory crohn's disease
JP2017524732A (en) * 2014-06-20 2017-08-31 ラトガース,ザ ステート ユニバーシティ オブ ニュー ジャージー How to treat lung inflammation

Also Published As

Publication number Publication date
US7199125B2 (en) 2007-04-03

Similar Documents

Publication Publication Date Title
AU2009272838B2 (en) Novel phenylimidazopyrazines
EP2307413B1 (en) Novel phenyl-imidazopyridines and pyridazines
CA2728683C (en) Novel phenylpyrazinones as kinase inhibitors
US6239133B1 (en) Imidazoquinoxaline protein tyrosine kinase inhibitors
US8293759B2 (en) Spiropiperidine beta-secretase inhibitors for the treatment of alzheimer's disease
US9045499B2 (en) Heteroaryl-substituted hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
WO2006006490A1 (en) Spirocyclic compound
US10851116B2 (en) Bicyclic amines as novel JAK kinase inhibitors
US20060069101A1 (en) Prodrugs of protein tyrosine kinase inhibitors
CA3105534C (en) Thiadiazole irak4 inhibitors
US7199125B2 (en) Spiro-cyclic compounds useful as anti-inflammatory agents
CN102015709A (en) Heterocyclic antiviral compounds
US20090170830A1 (en) Tricyclic Beta-Secretase Inhibitors for the Treatment of Alzheimer's Disease
US7375237B2 (en) Pyrrolizine compounds useful as anti-inflammatory agents
US6974815B2 (en) Hexahydro-benzimidazolone compounds useful as anti-inflammatory agents
US6673825B2 (en) Urazole compounds useful as anti-inflammatory agents
AU2021241502A1 (en) Potent and selective irreversible inhibitors of IRAK1
IL98428A (en) 3-(1,2,4-oxadiazolyl) imidazo [1,5-a] quinazolineamine and quinoxalineamine compounds, their preparation and pharmaceutical compositions containing them
AU2002303808A1 (en) Urazole compounds useful as anti-inflammatory agents
WO2022093820A1 (en) Substituted morpholine compounds

Legal Events

Date Code Title Description
AS Assignment

Owner name: CEREP SA, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LAUNAY, MICHELE;POTIN, DOMINIQUE;MAILLET, MAGALI JEANNINE BLANDINE;AND OTHERS;REEL/FRAME:016688/0049;SIGNING DATES FROM 20041029 TO 20041103

Owner name: BRISTOL-MYERS SQUIBB CO., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DHAR, T. G. MURALI;IWANOWICZ, EDWIN;REEL/FRAME:016666/0593;SIGNING DATES FROM 20041206 TO 20041207

STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

FPAY Fee payment

Year of fee payment: 8

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 12TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1553); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 12